EP1692113B1 - Aryl imidazoles and their use as anti-cancer agents - Google Patents

Aryl imidazoles and their use as anti-cancer agents Download PDF

Info

Publication number
EP1692113B1
EP1692113B1 EP04799154.2A EP04799154A EP1692113B1 EP 1692113 B1 EP1692113 B1 EP 1692113B1 EP 04799154 A EP04799154 A EP 04799154A EP 1692113 B1 EP1692113 B1 EP 1692113B1
Authority
EP
European Patent Office
Prior art keywords
compound
cancer
substituted
compounds
carcinoma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP04799154.2A
Other languages
German (de)
French (fr)
Other versions
EP1692113A1 (en
Inventor
Mario Huesca
Raed Al-Qawasmeh
Aiping H. Young
Yoon Lee
Lisa Lock
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aptose Bioscience Inc
Original Assignee
Lorus Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lorus Therapeutics Inc filed Critical Lorus Therapeutics Inc
Publication of EP1692113A1 publication Critical patent/EP1692113A1/en
Application granted granted Critical
Publication of EP1692113B1 publication Critical patent/EP1692113B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • This invention pertains to the field of anti-cancer compounds and, in particular, to the therapeutically active 2,4,5-trisubstituted imidazole compounds for use in the treatment of cancer.
  • a cancer is a malignant tumour of potentially unlimited growth. It is primarily the pathogenic replication (a loss of normal regulatory control) of various given types of cells found in the human body. By select mutation resulting from a primary lesion, the DNA of a cancer cell evolves and converts the cell into an autonomous system. Conventional cancer treatments have focused mainly on killing cancerous cells. Chemotherapeutic agents currently used for anti-cancer/anti-tumour therapy are selected for their toxicity towards rapidly proliferating cells. Most of them cause undesirable systemic effects such as cardiac or renal toxicity, marrow aplasia, alopecia, nausea and vomiting.
  • Heterocyclic compounds especially heterocyclic azole derivatives, have been shown to have a wide spectrum of biological activities.
  • One class of compounds with interesting biological activities is the imidazole (derivatives containing a five-membered heterocyclic azole).
  • a variety of biological activities have been reported for imidazole derivatives with different substitution patterns ( Lee et al. Nature 1994 327:739-745 ; Abdel-Meguid et al. Biochemistry, 1994, 33:11671 ; Heerding et al. Bioorg. Med. Chem. Lett. 2001, 11:2061-2065 ; Bu et al. Tetrahedron Lett. 1996, 37:7331-7334 ; Lewis JR. Nat. Prod. Rep. 1999, 16:389-418 ; Lewis JR. Nat. Prod. Rep. 1998, 15:417-437 and 371-395 ).
  • aryl-imidazole derivatives can act as modulators of multi-drug resistance in cancer cells ( Zhang et al. Bioorg. Med. Chem. Lett. 2000, 10:2603-2605 ), inhibitors of p38 MAP kinase ( Adams et al. Bioorg. Med. Chem. Lett. 2001, 11:867-2870 , McLay et. al. Bioorg. Med. Chem. 2001, 9:537-554 ) and of cytokines ( U.S. Patent Nos.
  • triaryl-imidazole compounds can act as inhibitors of p38 MAP kinase (for example, see LoGrasso et al. Biochemistry. 1997, 36:10422-10427 ) and as modulators of multi-drug resistance in cancer cells ( Sarshar et al. Bioorg. Med. Chem. Lett. 2000,10:2599-2601 ), however, the majority of the literature indicates that these compounds have found use mainly as colour producing reagents. ( U.S. Patent Nos.
  • NCI National Cancer Institute
  • NCI Yeast Anticancer Drug Screen The screen is based on the ability of candidate compounds to inhibit the growth of Saccharmyces cerevisiae strains that have mutations in genes related to cell cycle control and DNA repair damage.
  • Compounds are initially screened against a panel of six yeast strains at a single concentration (Stage0).
  • Compounds with activity in Stage0 are re-screened against the same panel at two concentrations (Stage1).
  • Stage2 Selected compounds with activity in Stage1 that also show selectivity are re-screened against a panel of 13 yeast strains at five concentrations (Stage2). Many of the results from the screening have been made available on the NCI/DTP website. The approach adopted in this screen is dependent on a candidate compound exerting its activity on certain cellular pathways (i.e. cell cycle control or DNA repair damage). The results generated by this type of screen, therefore, represent a very preliminary stage of screening for potential anti-cancer drugs and do not necessarily correlate with the ability of a compound to inhibit the growth of cancer cells in vitro or in vivo.
  • the NCI also provides an in vivo screening program to try to identify potential anti-cancer drugs (NCI In Vivo Anticancer Drug Screen). Many of the results from this screening program are also available from the NCI/DTP website.
  • EP 0812829 describes tri-substituted imidazole compounds, which are useful in the treatment of cancer through sensitization of multi-drug resistant cancer cells to chemotherapeutic agents. Other compounds are described in J. Indian Chem. Soc., 1979, 56, 620-624 .
  • a compound for use in the treatment of cancer having the structural formula: or a salt thereof, wherein:
  • Also disclosed herein is a method of inhibiting neoplastic cell growth or proliferation in a mammal comprising administering to said mammal a therapeutically effective amount of a compound selected from the compounds of general formula (IV) and (VII), or a salt thereof.
  • the present invention provides a class of 2,4,5-trisubstituted imidazole compounds and them for use as anti-cancer agents.
  • substituted lower alkyl refers to lower alkyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom of the lower alkyl moiety.
  • lower alkenyl refers to a straight chain or branched hydrocarbon of two to ten carbon atoms or a cyclic hydrocarbon of three to ten carbon atoms, having at least one carbon to carbon double bond.
  • substituted lower alkenyl refers to lower alkenyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl, alkenyl, alkynyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom to produce a stable compound.
  • lower alkynyl refers to a straight chain or branched hydrocarbon of two to ten carbon atoms having at least one carbon to carbon triple bond.
  • substituted lower alkynyl refers to lower alkynyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl, alkenyl, alkynyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom to produce a stable compound.
  • alkoxy refers to the group -OR, where R is lower alkyl, substituted lower alkyl, acyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heteroalkyl, heteroarylalkyl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, or substituted cycloheteroalkyl as defined below.
  • acyl refers to groups -C(O)R, where R is hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, cycloalkyl or substituted cycloalkyl.
  • aryloxy refers to groups -OAr, where Ar is an aryl, substituted aryl, heteroaryl, or substituted heteroaryl group as defined below.
  • amino refers to the group NRR', where R and R' may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, heteroaryl, cycloalkyl, or substituted heteroaryl as defined below, acyl, D or L aminoacid or a protected form thereof.
  • amido refers to the group -C(O)NRR', where R and R' may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, hetaryl, substituted hetaryl as defined below.
  • Carboxyl refers to the group -C(O)OR, where R may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, hetaryl, substituted hetaryl and the like as defined.
  • aryl refers to an aromatic carbocyclic group having at least one aromatic ring (e.g., phenyl or biphenyl) or multiple condensed rings in which at least one ring is aromatic, (e.g., 1,2,3,4-tetrahydronaphthyl, naphthyl, anthryl, phenanthryl, 9-fluorenyl, dibenzocycloheptatrienyl etc.).
  • heterocycle refers to a saturated, unsaturated, or aromatic carbocyclic group having a single ring (e.g., morpholino, pyridyl or furyl) or multiple condensed rings (e.g., naphthpyridyl, quinoxalyl, quinolinyl, indolizinyl, indanyl or benzo[b]thienyl) and having at least one hetero atom, such as N, O or S, within the ring.
  • a single ring e.g., morpholino, pyridyl or furyl
  • multiple condensed rings e.g., naphthpyridyl, quinoxalyl, quinolinyl, indolizinyl, indanyl or benzo[b]thienyl
  • hetero atom such as N, O or S
  • substituted heterocycle refers to heterocycle optionally substituted with, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl, substituted heteroaryl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido or cyano and the like.
  • heteroaryl or “hetaryl” refer to a heterocycle in which at least one heterocyclic ring is aromatic.
  • aralkyl refers to the group -R-Ar where Ar is an aryl group and R is lower alkyl or substituted lower alkyl group.
  • Aryl groups can optionally be unsubstituted or substituted with, e.g., halogen, lower alkyl, alkoxy, alkyl thio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • heteroalkyl refers to the group -R-Het where Het is a heterocycle group and R is a lower alkyl group.
  • Heteroalkyl groups can optionally be unsubstituted or substituted with e.g., halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • heteroarylalkyl refers to the group -R-HetAr where HetAr is an heteroaryl group and R lower alkyl or substituted loweralkyl.
  • Heteroarylalkyl groups can optionally be unsubstituted or substituted with, e.g., halogen, lower alkyl, substituted lower alkyl, alkoxy, alkylthio, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • cycloalkyl refers to a cyclic or polycyclic alkyl group containing 3 to 15 carbon.
  • these may be multiple condensed rings in which one of the distal rings maybe aromatic (e.g. tetrahydronaphthalene, etc.).
  • substituted cycloalkyl refers to a cycloalkyl group comprising one or more substituents with, e.g halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido or cyano and the like.
  • cycloheteroalkyl refers to a cycloalkyl group wherein one or more of the ring carbon atoms is replaced with a heteroatom (e.g., N, O, S or P).
  • substituted cycloheteroalkyl refers to a cycloheteroalkyl group as herein defined which contains one or more substituents, such as halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • substituents such as halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • alkyl cycloalkyl refers to the group -R-cycloalkyl where cycloalkyl is a cycloalkyl group and R is a lower alkyl or substituted lower alkyl.
  • Cycloalkyl groups can optionally be unsubstituted or substituted with e.g. halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • therapy and treatment refer to an intervention performed with the intention of alleviating the symptoms associated with, preventing the development of, or altering the pathology of a disease, disorder or condition.
  • therapy and treatment are used in the broadest sense, and include the prevention (prophylaxis), moderation, reduction, and curing of a disease, disorder or condition at various stages.
  • Those in need of therapy/treatment include those already having the disease, disorder or condition as well as those prone to, or at risk of developing, the disease, disorder or condition and those in whom the disease, disorder or condition is to be prevented.
  • subject refers to an animal in need of treatment.
  • animal refers to both human and non-human animals, including, but not limited to, mammals, birds and fish.
  • Administration of the compounds of the invention "in combination with" one or more further therapeutic agents is intended to include simultaneous (concurrent) administration and consecutive administration. Consecutive administration is intended to encompass various orders of administration of the therapeutic agent(s) and the compound(s) of the invention to the subject.
  • the term "about” refers to a +/-10% variation from the nominal value. It is to be understood that such a variation is always included in any given value provided herein, whether or not it is specifically referred to.
  • the present invention includes the compound of the structural formula, for use in the treatment of cancer: or a salt thereof, wherein:
  • the present invention includes pharmaceutically acceptable salts of the compounds defined by Formula VI.
  • Compounds according to the present invention can possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with a number of organic and inorganic bases, and organic and inorganic acids, to form pharmaceutically acceptable salts.
  • pharmaceutically acceptable salt refers to a salt of a compound of Formula VI, which is substantially non-toxic to living organisms.
  • Typical pharmaceutically acceptable salts include those salts prepared by reaction of the compound of the present invention with a pharmaceutically acceptable mineral or organic acid or an organic or inorganic base. Such salts are known as acid addition and base addition salts.
  • Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, phosphoric acid, and the like, and organic acids such as p -toluenesulphonic acid, methanesulphonic acid, oxalic acid, p -bromophenylsulphonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, phosphoric acid, and the like
  • organic acids such as p -toluenesulphonic acid, methanesulphonic acid, oxalic acid, p -bromophenylsulphonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • salts examples include the sulphate, pyrosulphate, bisulphate, sulphite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, hydrochloride, dihydrochloride, isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1,4-dioate, hexyne-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, hydroxybenzoate, methoxybenzoate, phthalate, xylenesulphonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, gamma
  • Salts of amine groups may also comprise quarternary ammonium salts in which the amino nitrogen carries a suitable organic group such as an alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, or aralkyl moiety.
  • Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like.
  • Bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, calcium hydroxide, calcium carbonate, and the like.
  • solvates of a compound of Formula VI are pharmaceutically acceptable solvates of a compound of Formula VI.
  • solvents such as water, methanol, ethanol and acetonitrile to form pharmaceutically acceptable solvates such as the corresponding hydrate, methanolate, ethanolate and acetonitrilate.
  • the compounds of the present invention may have multiple asymmetric (chiral) centres. As a consequence of these chiral centres, the compounds of the present invention occur as racemates, mixtures of enantiomers and as individual enantiomers, as well as diastereomers and mixtures of diastereomers. All asymmetric forms, individual isomers and combinations thereof, are within the scope of the present invention.
  • Non-toxic metabolically-labile esters or amides of a compound of Formula VI are those that are hydrolysed in vivo to afford the compound of Formula VI and a pharmaceutically acceptable alcohol or amine.
  • Examples of metabolically-labile esters include esters formed with (1-6C) alkanols, in which the alkanol moiety may be optionally substituted by a (1-8C) alkoxy group, for example methanol, ethanol, propanol and methoxyethanol.
  • Non-limiting examples of metabolically-labile amides include amides formed with amines such as methylamine.
  • triaryl imidazole compounds can be prepared by a number of standard techniques. Compounds, therefore, can be prepared by several general synthetic methods, for example, as described by Grimmett, ( Grimmett, M.R., Comprehensive Heterocyclic Chemistry: The Structure, Reaction, Synthesis and Uses of Heterocyclic Compounds, A. R. Katrizky and C. W. Rees, eds., Vol. 5, Pergamon Press. Oxford, 1984, pp. 457-498 ; Grimmett, M. R., Imidazole and Benzimidazole Synthesis, Academic Press, San Diego CA, 1997 ).
  • the compounds of Formula (XXXI) and (XXXII) are either commercially available or may be prepared using standard procedures known to a person skilled in the relevant art. Compounds of Formula (XXXI), therefore, can be prepared by several general synthetic methods, for example, as described by: Fischer et. al (J. Am. Chem. Soc. 1961, 83, 4208-4210 ); Guijarro et al. (J. Am. Chem. Soc. 1999, 121, 4155-4157 ); Chi et. al. (Synth. Comm. 1994, 24(15), 2119-2122 ) and Armesto et. al. (Synthesis, 1988, 799-801 ).
  • a candidate compound of Formula VI to inhibit neoplastic cell growth and/or proliferation can be tested using standard techniques known in the art.
  • compounds of Formula VI that demonstrate inhibitory activity may be further tested in vitro and/or in vivo in combination with various known chemotherapeutics to evaluate their potential use in combination therapies.
  • Exemplary methods of testing candidate compounds are provided below and in the Examples included herein. One skilled in the art will understand that other methods of testing the compounds are known in the art and are also suitable for testing candidate compounds.
  • Candidate compounds of the invention can be assayed initially in vitro for their ability to inhibit cell growth (i.e. their cytotoxicity) using standard techniques.
  • cells of a specific test cell line typically a cancer cell line
  • a suitable density e.g . approximately 1 x 10 4
  • cell survival is assessed, for example, by assaying for tetrazolium salt (or modified tetrazolium salt) cleavage, or by using the resazurin reduction test (see Fields & Lancaster (1993) Am. Biotechnol. Lab.
  • Inhibition of cell growth is determined by comparison of cell survival in the treated culture with cell survival in one or more control cultures, for example, cultures not pre-treated with the candidate compound and/or those pre-treated with a control compound (typically a known therapeutic).
  • a control compound typically a known therapeutic.
  • Other suitable techniques for assessing cytotoxicity are known in the art.
  • Assays that measure metabolic activity can also be used to assess the effect of candidate compounds on cell activation and /or proliferation, due the fact that proliferating cells are metabolically more active than resting cells.
  • Candidate compounds can also be tested in vitro for their ability to inhibit anchorage-independent growth of tumour cells.
  • Anchorage-independent growth is known in the art to be a good indicator of tumourigenicity.
  • anchorage-independent growth is assessed by plating cells from an appropriate cancer cell-line onto soft agar and determining the number of colonies formed after an appropriate incubation period. Growth of cells treated with the candidate compound can then be compared with that of cells treated with an appropriate control (as described above).
  • in vitro testing of the candidate compounds is conducted in a human cancer cell-line.
  • suitable human cancer cell-lines for in vitro testing of the compounds of the present invention include, but are not limited to, colon and colorectal carcinoma cell lines such as HT-29, CaCo, LoVo, COLO320 and HCT-116; non small cell lung cancer cell lines such as NCI-H460, small cell lung cancer cell lines such as H209; breast cancer cell lines such as MCF-7, T47D and MDA-MB-231; ovarian cancer cell lines such as SK-OV-3; prostate cancer cell lines such as PC-3 and DU-145; chronic myeloid leukemia cell lines such as K562; bladder cancer cell lines such as T24; brain cancer cell lines such as U-87-MG; pancreatic cancer cell lines such as AsPC-1, SU.86.86 and BxPC-3; kidney cancer cell lines such as A498 and Caki-1; liver cancer cell
  • the selectivity of the candidate compounds of the invention may also be tested, i.e. the ability of the compound to demonstrate some level of selective action toward neoplastic (or cancer) cells in comparison to normal proliferating cells.
  • An exemplary method of assessing the differential sensitivity between normal and cancer cells for a compound has been described by Vassilev et al. (Anti-Cancer Drug Design (2001) 16:7 ). This method involves the comparison of IC 90 values, i.e. the molar concentration of a test compound required to cause 90% growth inhibition of exponentially growing cells.
  • the IC 90 values for candidate compounds can be evaluated in various cancer cell lines (such as those outlined above) and normal cells (such as HUVEC and/or WI38 cells) and compared. IC 90 values can be measured using a variety of standard techniques including those described above for cytotoxicity testing.
  • assays to investigate potential mechanisms of action of the compounds may be conducted if desired in order to provide information useful in determining what aspects of tumour growth the compounds affect. This type of information may help to determine cancer types that will benefit from treatment with the compounds.
  • assays include, but are not limited to, cell-cycle analysis (for example, employing flow cytometry techniques), apoptosis assays (such as DNA fragmentation analysis), anti-angiogenesis assays (for example, various Matrigel assays, including cord formation and Matrigel plug assays) and immunohistochemical analysis.
  • Toxicity of the candidate compounds can also be initially assessed in vitro using standard techniques.
  • human primary fibroblasts can be treated in vitro with a compound of the invention and then tested at different time points following treatment for their viability using a standard viability assay, such as the assays described above or the trypan-blue exclusion assay.
  • Cells can also be assayed for their ability to synthesize DNA, for example, using a thymidine incorporation assay, and for changes in cell cycle dynamics, for example, using a standard cell sorting assay in conjunction with a fluorocytometer cell sorter (FACS).
  • FACS fluorocytometer cell sorter
  • the ability of the candidate compounds to inhibit tumour growth, proliferation and/or metastasis in vivo can be determined in an appropriate animal model using standard techniques known in the art (see, for example, Enna, et al., Current Protocols in Pharmacology, J. Wiley & Sons, Inc., New York, NY ). Exemplary protocols are provided below and in the Examples. Non-limiting examples of suitable animal models are provided in Table 1.
  • current animal models for screening anti-tumour compounds are xenograft models, in which a human tumour has been implanted into an animal.
  • the candidate compounds can be tested in vivo on solid tumours using mice that are subcutaneously grafted or injected with 30 to 60 mg of a tumour fragment, or an appropriate number of tumour cells ( e.g . about 10 6 to 10 7 ) on day 0.
  • the animals bearing tumours are mixed before being subjected to the various treatments and controls.
  • tumours are allowed to: develop to the desired size, animals having insufficiently developed tumours being eliminated.
  • the selected animals are distributed at random to undergo the treatments and controls.
  • tumour-bearing animals may also be subjected to the same treatments as the tumour-bearing animals in order to be able to dissociate the toxic effect from the specific effect on the tumour.
  • Chemotherapy generally begins from 3 to 22 days after grafting, depending on the type of tumour, and the animals are observed every day.
  • Candidate compounds can be administered to the animals, for example, by bolus infusion.
  • the different animal groups are weighed about 3 or 4 times a week until the maximum weight loss is attained, after which the groups are weighed at least once a week until the end of the trial.
  • tumours are measured about 2 or 3 times a week until the tumour reaches a pre-determined size and / or weight, or until a pre-determined time period has passed, or until the animal dies (if this occurs before the tumour reaches the pre-determined size / weight).
  • the animals are then sacrificed and the tissue histology, size and / or proliferation of the tumour assessed.
  • tumours into animals i.e . the implantation of cancer cells of a certain type into the corresponding tissue in the animal, such as pancreatic cancer cells into the pancreas
  • pancreatic cancer cells into the pancreas may also be used to assess the effect of the candidate compounds on tumour growth and proliferation.
  • the effect of the candidate compound on spontaneous tumours in normal mice can be assessed.
  • the effect of the candidate compounds on drug-resistant tumours can be assessed in vivo by utilising a drug- or multidrug-resistant cancer cell in the xenograft experiments.
  • the animals are grafted or injected with a particular number of cells, and the anti-tumour activity is determined by the increase in the survival time of the treated mice relative to the controls.
  • tumour cells are typically treated with the compound ex vivo and then injected into a suitable test animal. The spread of the tumour cells from the site of injection is then monitored over a suitable period of time.
  • test animals would be treated with both the chemotherapeutic agent and the candidate compound of the invention.
  • Control animal could include animals treated with the chemotherapeutic alone, animals treated with the candidate compound alone and/or untreated animals.
  • In vivo toxic effects of the compounds of the invention can be evaluated by standard techniques, for example, by measuring their effect on animal body weight during treatment and by performing haematological profiles and liver enzyme analysis after the animal has been sacrificed (survival assays).
  • Table I Examples of in vivo models of human cancer Cancer Model Cell Type Tumour Growth Assay Prostate (PC-3, DU145) Human solid tumour xenografts in mice (sub-cutaneous injection) Breast (MDA-MB-231, MVB-9) Colon (HT-29) Lung (NCI-H460, NCI-H209) Pancreatic (ASPC-1, SU86.86) Pancreatic: drug resistant (BxPC-3) Skin (A2058, C8161) Cervical (SIHA, HeLa-S3) Cervical: drug resistant (HeLa S3- HU-resistance) Liver (HepG2) Brain (U87-MG) Renal (Caki-1, A498) Ovary (SK-OV-3) Bladder (T24
  • Toxicity tests for potential drugs are well-known in the art (see, for example, Hayes, A.W., ed., (1994), Principles and Methods of Toxicology, 3rd ed., Raven Press, NY ; Maines, M., ed., Current Protocols in Toxicology, John Wiley & Sons, Inc., NY ).
  • In vitro acute toxicity testing of a compound of the invention can be performed using mammalian cell lines (see, for example, Ekwall, B., Ann. N.Y. Acad. Sci., (1983) 407:64-77 ). Selection of an appropriate cell line is dependent on the potential application of the candidate compound and can be readily determined by one skilled in the art.
  • In vivo toxicity testing can be performed by standard methodology. For example, by injecting varying concentrations of the candidate compound into an appropriate animal model. The compound can be injected once, or administration can be repeated over several days. The toxic effects of the compound can be evaluated over an appropriate time period by monitoring the general health and body weight of the animals. After the completion of the period of assessment, the animals can be sacrificed and the appearance and weight of the relevant organs determined. An indication of the toxicity of a compound can also be obtained during the in vivo anti-cancer testing of the compound.
  • the compounds of the invention can be used in the treatment and/or stabilisation of various types of cancers.
  • the compounds may exert either a cytotoxic or cytostatic effect resulting in a reduction in the size of a tumour, the slowing or prevention of an increase in the size of a tumour, an increase in the disease-free survival time between the disappearance or removal of a tumour and its reappearance, prevention of an initial or subsequent occurrence of a tumour ( e.g. metastasis), an increase in the time to progression, reduction of one or more adverse symptom associated with a tumour, or an increase in the overall survival time of a subject having cancer.
  • the compounds can be used alone or they can be used as part of a multi-drug regimen in combination with one or more known therapeutics.
  • One embodiment of the present invention provides for the use of the compounds of Formula VI in the treatment and/or stabilisation of a solid tumour.
  • leukaemia refers broadly to progressive, malignant diseases of the blood-forming organs. Leukaemia is typically characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow but can also refer to malignant diseases of other blood cells such as erythroleukaemia, which affects immature red blood cells. Leukaemia is generally clinically classified on the basis of (1) the duration and character of the disease - acute or chronic; (2) the type of cell involved - myeloid (myelogenous), lymphoid (lymphogenous) or monocytic, and (3) the increase or non-increase in the number of abnormal cells in the blood - leukaemic or aleukaemic (subleukaemic).
  • Leukaemia includes, for example, acute nonlymphocytic leukaemia, chronic lymphocytic leukaemia, acute granulocytic leukaemia, chronic granulocytic leukaemia, acute promyelocytic leukaemia, adult T-cell leukaemia, aleukaemic leukaemia, aleukocythemic leukaemia, basophylic leukaemia, blast cell leukaemia, bovine leukaemia, chronic myelocytic leukaemia, leukaemia cutis, embryonal leukaemia, eosinophilic leukaemia, Gross' leukaemia, hairy-cell leukaemia, hemoblastic leukaemia, hemocytoblastic leukaemia, histiocytic leukaemia, stem cell leukaemia, acute monocytic leukaemia, leukopenic leukaemia, lymphatic leukaemia, lymphoblastic leukaemia, lymphocytic leuk
  • tumour generally refers to a tumour which originates in connective tissue, such as muscle, bone, cartilage or fat, and is made up of a substance like embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance.
  • Sarcomas include soft tissue sarcomas, chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abemethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumour sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, granulocytic sarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented haemorrhagic
  • carcinoma refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases.
  • exemplary carcinomas include, for example, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colorectal carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiermoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex
  • carcinomas that originate in cells that make organs which have glandular (secretory) properties or that originate in cells that line hollow viscera, such as the gastrointestinal tract or bronchial epithelia. Examples include, but are not limited to, adenocarcinomas of the breast, lung, colon, pancreas and prostate.
  • the cancer to be treated or stabilized may be indolent or it may be aggressive.
  • the compounds of the invention can be used to treat refractory cancers, advanced cancers, recurrent cancers and metastatic cancers.
  • refractory cancers advanced cancers
  • recurrent cancers recurrent cancers
  • metastatic cancers One skilled in the art will appreciate that many of these categories may overlap, for example, aggressive cancers are typically also metastatic.
  • Advanced cancer refers to overt disease in a patient that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy. Advanced disease may refer to a locally advanced cancer or it may refer to metastatic cancer.
  • metastatic cancer refers to cancer that has spread from one part of the body to another.
  • the compounds of Formula VI as "sensitizing agents," which selectively inhibit the growth of cancer cells.
  • the compound alone does not have a cytotoxic effect on the cancer cell, but provides a means of weakening the cancer cells, and thereby facilitates the benefit from conventional anti-cancer therapeutics.
  • an anti-cancer therapeutic is a compound, composition or treatment that prevents or delays the growth and/or metastasis of cancer cells.
  • anti-cancer therapeutics include, but are not limited to, chemotherapeutic drug treatment, radiation, gene therapy, hormonal manipulation, immunotherapy and antisense oligonucleotide therapy.
  • chemotherapeutic drugs are known in the art and can be used in combination therapies with a compound of the present invention.
  • chemotherapeutic drugs include broad spectrum chemotherapeutics, i. e. those that are useful in the treatment of a range of cancers, such as doxorubicin, capecitabine, mitoxantrone, irinotecan (CPT-11), cisplatin and gemcitabine.
  • broad spectrum chemotherapeutics i. e. those that are useful in the treatment of a range of cancers, such as doxorubicin, capecitabine, mitoxantrone, irinotecan (CPT-11), cisplatin and gemcitabine.
  • chemotherapeutic agents include, but are not limited to, hydroxyurea, busulphan, carboplatin, chlorambucil, melphalan, cyclophosphamide, Ifosphamide, danorubicin, epirubicin, vincristine, vinblastine, Navelbine® (vinorelbine), etoposide, teniposide, paclitaxel, docetaxel, cytosine, arabinoside, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C and the like.
  • the compounds of the invention are also suitable for use with standard combination therapies employing two or more chemotherapeutic agents. It is to be understood that anti-cancer therapeutics for use in the present invention also include novel compounds or treatments developed in the future.
  • the compounds of the present invention are typically formulated prior to administration.
  • the present invention thus provides pharmaceutical compositions comprising one or more compounds of the invention and a pharmaceutically acceptable carrier, diluent, or excipient.
  • the pharmaceutical compositions are prepared by known procedures using well-known and readily available ingredients.
  • Pharmaceutical compositions comprising one or more compounds of the invention in combination with one or more known cancer chemotherapeutics are also contemplated by the present invention.
  • Compounds of the invention or pharmaceutical compositions comprising the compounds may be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • the active compound is incorporated into an acceptable vehicle to form a composition for topical administration to the affected area, such as hydropohobic or hydrophilic creams or lotions, or into a form suitable for oral, rectal or parenteral administration, such as syrups, elixirs, tablets, troches, lozenges, hard or soft capsules, pills, suppositiories, oily or aqueous suspensions, dispersible powders or granules, emulsions, injectables, or solutions.
  • parenteral as used herein includes subcutaneous injections, intradermal, intra-articular, intravenous, intramuscular, intravascular, intrasternal, intrathecal injection or infusion techniques.
  • compositions intended for oral use may be prepared in either solid or fluid unit dosage forms.
  • Fluid unit dosage form can be prepared according to procedures known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • An elixir is prepared by using a hydroalcoholic (e.g., ethanol) vehicle with suitable sweeteners such as sugar and saccharin, together with an aromatic flavoring agent.
  • Suspensions can be prepared with an aqueous vehicle with the aid of a suspending agent such as acacia, tragacanth, methylcellulose and the like.
  • Solid formulations such as tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate: granulating and disintegrating agents for example, corn starch, or alginic acid: binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc and other conventional ingredients such as dicalcium phosphate, magnesium aluminum silicate, calcium sulfate, starch, lactose, methylcellulose, and functionally similar materials.
  • inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate
  • granulating and disintegrating agents for example, corn starch, or alginic acid: binding agents, for example starch, ge
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate maybe employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • Soft gelatin capsules are prepared by machine encapsulation of a slurry of the compound with an acceptable vegetable oil, light liquid petrolatum or other inert oil.
  • Aqueous suspensions contain active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxylmethylcellulose, methyl cellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia: dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example hepta-decaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoo
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n -propyl- p -hydroxy benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n -propyl- p -hydroxy benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example peanut oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavouring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol,
  • compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oil phase may be a vegetable oil, for example olive oil or peanut oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or a suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Adjuvants such as local anaesthetics, preservatives and buffering agents can also be included in the injectable solution or suspension.
  • compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter and polyethylene glycols.
  • compositions and methods of preparing pharmaceutical compositions are known in the art and are described, for example, in “Remington: The Science and Practice of Pharmacy” (formerly “ Remingtons Pharmaceutical Sciences”); Gennaro, A., Lippincott, Williams & Wilkins, Philidelphia, PA (2000 ).
  • Compounds of the invention may be administered to a subject by a variety of routes depending on the cancer to be treated, for example, the compounds may be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations.
  • the compounds are administered systemically to a subject, for example, by bolus injection or continuous infusion into a subject's bloodstream or by oral administration.
  • the compounds can be administered prior to, or after, administration of the chemotherapeutic agents, or they can be administered concomitantly.
  • the one or more chemotherapeutic may also be administered systemically, for example, by bolus injection, continuous infusion, or oral administration.
  • the compounds of the invention may be used as part of a neo-adjuvant therapy (to primary therapy), or as part of an adjuvant therapy regimen, where the intention is to cure the cancer in a subject.
  • the present invention contemplates the use of the compounds of at various stages in tumour development and progression, including in the treatment of advanced and/or aggressive neoplasias (i.e . overt disease in a subject that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy), metastatic disease, locally advanced disease and/or refractory tumours ( i.e. a cancer or tumour that has not responded to treatment).
  • Primary therapy refers to a first line of treatment upon the initial diagnosis of cancer in a subject.
  • exemplary primary therapies may involve surgery, a wide range of chemotherapies and radiotherapy.
  • adjuvant therapy refers to a therapy that follows a primary therapy and that is administered to subjects at risk of relapsing. Adjuvant systemic therapy is usually begun soon after primary therapy to delay recurrence, prolong survival or cure a subject.
  • the compounds of the invention can be used alone or in combination with one or more other chemotherapeutic agents as part of a primary therapy or an adjuvant therapy.
  • Combinations of the compounds of the invention and standard chemotherapeutics may act to improve the efficacy of the chemotherapeutic and, therefore, can be used to improve standard cancer therapies.
  • This application can be important in the treatment of drug-resistant cancers which are not responsive to standard treatment. Drug-resistant cancers can arise, for example, from heterogeneity of tumour cell populations, alterations in response to chemotherapy and increased malignant potential. Such changes are often more pronounced at advanced stages of disease.
  • the dosage to be administered is not subject to defined limits, but it will usually be an effective amount. It will usually be the equivalent, on a molar basis of the pharmacologically active free form produced from a dosage formulation upon the metabolic release of the active free drug to achieve its desired pharmacological and physiological effects.
  • the compositions may be formulated in a unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Examples of ranges for the compound(s) in each dosage unit are from about 0.05 to about 100 mg, or more usually, from about 1.0 to about 50 mg.
  • Daily dosages of the compounds of the present invention will typically fall within the range of about 0.01 to about 100 mg/kg of body weight, in single or divided dose.
  • the actual amount of the compound(s) to be administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.
  • the above dosage range is given by way of example only and is not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing harmful side effects, for example, by first dividing the larger dose into several smaller doses for administration throughout the day.
  • kits containing one or more compounds of Formula VI for use in the treatment of cancer can be lyophilized and the kit can additionally contain a suitable solvent for reconstitution of the lyophilized components.
  • Individual components of the kit would be packaged in separate containers and, associated with such containers, can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, for use or sale for human or animal administration.
  • the liquid solution can be an aqueous solution, for example a sterile aqueous solution.
  • the compounds may be formulated into a pharmaceutically acceptable syringeable composition.
  • the container means may itself be an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the formulation may be applied to an infected area of the subject, such as the lungs, injected into an subject, or even applied to and mixed with the other components of the kit.
  • Pharmaceutical kits or packs comprising one or more compound of the present invention in combination with one or more standard chemotherapeutic for combination therapy applications are also contemplated by the present invention.
  • the suspension solid was then filtered and the crude solid was dissolved in ethyl acetate, dried over sodium sulfate and filtered, the organic solvent was removed by vacuum.
  • the products was then either recrystalized with alcohol or separated by column chromatography using petroleum ether-Ethyl acetate as an eluant.
  • Selected compounds of the invention were tested for anti-cancer activity in vitro using a human colon carcinoma cells (HT-29) and human non-small cell lung cancer cells (H460).
  • the cells were maintained in ⁇ -MEM medium (Wisent, St-Bruno, Qc) supplemented with 10% FBS, and grown at 37°C in an atmosphere of 5% CO 2 .
  • Cells were transferred onto 150mm tissue culture plates and grown until sub-confluency (70-80%) prior to their use.
  • the anti-cancer activity in vitro was evaluated by a cell proliferation assay based on the ability of live cells to reduce the tetrazolium salt XTT to orange coloured compounds of formazan (XTT cell proliferation kit II, Roche Applied Science, Montreal, QC).
  • HT-29 colon cancer cells
  • NCI-H460 non-small cell lung cancer cells
  • Examples 94, 97, 99, 100, 101, 102, 103, 104, 105 and 106 are in accordance with the present invention.
  • Cells were maintained in ⁇ -MEM medium (Wisent, St-Bruno, QC) supplemented with 10% FBS, and grown at 37°C in an atmosphere of 5% CO 2 . They were transferred onto 150mm tissue culture plates and grown until sub-confluency (70-80%) prior to their use.
  • the anti-cancer activity in vitro was evaluated by a cell proliferation assay based in the ability of live cells to reduce the tetrazolium salt XTT to orange coloured compounds of formazan (XTT cell proliferation kit II, Roche Applied Science, Montreal, QC).
  • the following cancer cell lines were tested: HT-29 colon carcinoma, A498 renal carcinoma, Caki-1 renal carcinoma, C8161 melanoma, MDA-MB-231 breast adenocarcinoma, A2058 metastatic melanoma, SK-OV-3 ovarian adenocarcinoma, Hep G2 liver carcinoma, AsPC-1 pancreatic adenocarcinoma, PC3 metastatic prostate adenocarcinoma.
  • WI 38 is a human lung fibroblast cell line.
  • Figure 1 depicts the results with compound 92; Figure 2 depicts the results with compound 28; Figure 3 depicts the results with compound 50; and Figure 4 depicts the results with compound 42.
  • Example 51 Concentration Dependence of Inhibition of Cancer Cell Proliferation by Compound 45 in vitro
  • Example 50 The effect of various concentrations of compound 45 on various cancer cell lines was tested following the general protocol outlined in Example 50, with the following exceptions.
  • Cell survival was assessed 48 h, 72 h and 6 days post-treatment by incubating cells with XTT for 2 h.
  • the cancer cell lines utilised in this example were the same as those listed in Example 50, together with the cervical carcinoma cell line KB.
  • Figures 5 and 6 depict cell survival after treatment with various concentrations of compound 45. A. 48 h after treatment, B. 72 h after treatment and C. 6 days after treatment.
  • Example 52 Concentration Dependence of Inhibition of Cancer Cell Proliferation by Compounds 45 and 99 In Vitro
  • the NCI conducts a standard 48/72 hour 60 cell line assay and an in vitro time course assay as described in Alley et al. ( Cancer Res (1988) 48:589).
  • a standard 60 cell line assay a minimum of 5 concentrations of the test compound are tested at 10-fold dilutions against 60 cell lines and cell growth is assayed at 48 and 72 hours using a sulphorhodamine B assay.
  • tumour cells are treated with the test compound at various time points, then washed and grown in medium free of the test compound until the end of the experiment at 144 hrs.
  • This assay employs 20% FBS to better approximate the minimum c x t (concentrations and times) test compound exposure conditions that are required to achieve activity in vivo.
  • Cell growth is quantified by an MTT assay (similar to the XTT assay described above) and the concentration of the test compound required for growth inhibition is determined.
  • the inhibitory effect of the test compounds are expressed as a GI 50 value, which represents the molar concentration of the test compound that results in 50% growth inhibition.
  • Example 55 The above compounds (as shown in Example 55), together with those shown below, were tested for their ability to inhibit the proliferation of HT-29 colon carcinoma cells in vitro.
  • the protocol described in Example 50 was utilised with the exception that cell survival was assessed after either 2 or 6 days of treatment.
  • Each compound was tested at concentrations of 0.2, 2, 10 and 25 ⁇ M (compounds 110, 30, 101,113, 103, 107, 108 and 109 ) or at concentrations of 2.5, 10 and 25 ⁇ M (compounds 112, 114, 78, 111 and 45 ).
  • the results are shown in Figures. 12 and 13 .
  • the results shown in Figure 12 reflect cell survival 6 days after treatment with the listed compounds.
  • Figure 13A shows cell survival 2 days after treatment with the listed compounds
  • Figure 13B shows cell survival 6 days after treatment.
  • Compounds 101, 113, 45 and 114 are compounds of the invention.
  • Example and the following Example 58 describe in vivo efficacy studies of various compounds of the invention performed using a mouse xenograft model using the human colon adenocarcinoma cell line HT-29. The following compounds were tested (Compounds 44, 45 and 46 are compounds of the invention).
  • the treatment schedule consisted of 2 x 200 ⁇ l intraperitoneal injections per day of 5 mg/ml (100 mg/Kg/d) for five days and 2 days break, for 4 weeks. Tumour sizes were measured during the course of the treatment using calipers, mice were then sacrificed by cervical dislocation and tumours surgically removed and weighed.
  • Figures 14 shows the average tumour size (mm 3 ) in the different groups of mice.
  • Figures 15 and 16 show the average tumour weight per group of mice and per individual mouse, respectively.
  • FIG. 17 depicts the average tumour size (mm 3 ) in the different groups of mice.
  • Compound 45 was tested for its ability to function as a kinase inhibitor using the kinase profiler service from Upstate Biotechnologies.
  • the general protocol employed is as follows: recombinant kinases were incubated with specific substrates, 10 mM MgAcetate, and [ ⁇ - 33 P-ATP]. The reaction was initiated by the addition of MgATP mix. After incubation at room temperature for 40 minutes, the reaction was stopped by the addition of 5 ⁇ l of a 3% phosphoric acid solution. 10 ⁇ l of the reaction was then spotted on to a P30 filtermat and washed 3 times for 5 min. in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
  • PI 3-kinase-y (PI3K- ⁇ ) activity was determined with the PIProfilerTM assay, which measures the binding of the GRP1 pleckstrin homology (PH) domain to PIP3, the product of PI3K acting on its physiological substrate PIP2.
  • Compound 45 is intrinsically fluorescent, which allowed the subcellular localization of this compound to be examined by fluorescent microscopy. Fluorescent microscopy was performed at the Microscopy Imaging Centre, Faculty of Medicine, University of Toronto. Cells were treated with 100 ⁇ M of compound 45 ( Figure 20A , B, D, E) or 1 ⁇ M doxorubicin ( Figure 20C ) for 1 hour, washed once in PBS, fixed in 3.7% formaldehyde/PBS for 10 minutes, washed three times in PBS and mounted with Immuno-fluoro. Images were obtained with a Zeiss laser scanning fluorescent microscope with an excitation filter range of 360-370 nm (compound 45) or 530-560 nm (doxorubicin). For Figure 20B and C , differential interference contrast images were overlaid with fluorescent images.
  • Compound 45 localizes to punctuate spots in the perinuclear area of HT-29 colon adenocarcinoma cells ( Figure 20A ), and is excluded from the nucleus and plasma membrane regions ( Figure 20B ).
  • the anti-cancer agent, doxorubicin which is also intrinsically fluorescent, is localized in the nucleus ( Figure 20C ).
  • a similar localization for compound 45 was observed in A498 renal carcinoma cells ( Figure 20D ) and C8161 melanoma cells ( Figure 20E ).
  • FIG. 21 shows differential interference contrast (DIC) images (top row) and fluorescent images (lower row) of the same cells stained with DAPI, a. cell permeable marker for the nucleus.
  • DIC differential interference contrast
  • Compounds of the invention that demonstrate the ability to decrease the growth or proliferation of at least one cancer cell line may undergo further testing to evaluate their selectivity towards cancer cells.
  • An exemplary method to measure the selectivity of the compounds of the present invention is provided below.
  • IC 90 values of selected compounds on a panel of normal actively proliferating cells (HUVEC and WI38) and cancer cells representing colon (HT-29), lung (NCI-H460), breast (MDA-MB-231) and prostate cancer (PC-3) are measured.
  • Compounds with 2-fold or higher overall selectivity to the panel of cancer cell lines at IC 90 are identified as potential therapeutics.
  • IC 90 values are determined using the XTT assay as an indicator of growth arrest and/or cytotoxicity. This assay is conducted as outlined in Example 50. Percentage inhibition is calculated for each cell line and IC 90 values for each compound and cell type determined. The average IC 90 values for the normal cells are calculated and divided by the average IC 90 values for the cancer cell lines. Compounds with a selectivity ratio of >2 are identified and chosen for further optimization and/or testing.
  • mice models that can be utilized to investigate the efficacy of selected compounds include, but are not limited to, xenografts of various human tumour types, inoculated subcutaneously into nude mice or mice with severe combined immunodeficiency disorder (SCID) as described above; orthotopic implantation of various human tumours in nude or SCID mice for investigation of effects on the tumour in the target organ (for example, a pancreatic cancer cell graft implanted directly into the pancreas of the animal), and investigation of spontaneous tumours in normal mice.
  • SCID severe combined immunodeficiency disorder
  • a selected compound in order to provide evidence of the efficacy of a selected compound as a single agent, it may be evaluated, for example, in specific models (xenograft or orthotopic) for representative human cancers such as pancreas, skin (melanoma), kidney, colon, breast, lung, liver, ovary, prostate, bladder and brain. Similar studies can be conducted to evaluate the performance of test compounds in combination with other standard therapeutic modalities used in the treatment of human cancers.
  • mice For typical xenograft studies, 5-6 week old, female, CD-1 athymic nude mice, (Charles River, Montreal, QC) are acclimatized in a pathogen-free facility for at least 1 week. Animal protocols followed are in compliance with the Guide for the Care and Use of Laboratory Animals in Canada. Approximately 10 6 -10 7 human tumour cells in 100 ml PBS are subcutaneously injected into the right flank of each mouse. Once tumours reach an approximate volume of 100 mm 3 (several days post tumour cell injection), mice are randomized by tumour size into control and treatment groups. Test compounds are administered at various doses 5 days a week for several weeks. Control animals receive vehicle alone (negative control) and/or a standard chemotherapeutic (positive control) for the same period.
  • tumour dimensions length, width, and height
  • Tumour volume is calculated by the formula L x W x H/2, where L indicates length, W indicates width and H indicates height.
  • L indicates length
  • W indicates width
  • H indicates height.
  • the mice are sacrificed when the tumour burden reaches approximately 10% of total body weight and excised tumours are weighed.
  • a standard bar graph is used to demonstrate the differences in tumour weights with each bar representing mean tumour weight.
  • the potential mechanism of action of selected compounds can be investigated using assays such as cell-cycle analysis, apoptosis assays, anti-angiogenesis assays and immunohistochemical analysis.
  • assays such as cell-cycle analysis, apoptosis assays, anti-angiogenesis assays and immunohistochemical analysis.
  • a representative example of each type of assay is provided below.
  • Alterations in cell cycle are determined using flow cytometric analyses.
  • Tumour cells sensitive to a test compound are synchronized by plating in medium containing 0.5% FBS for 24 h followed by culturing in FBS-free medium for 48 h. The cells are then released into complete medium containing 0.1% DMSO (vehicle control) or the test compound at an appropriate concentration (e.g. 3 x IC 90 value), harvested 16 to 24 h following treatment, washed twice with cold PBS and fixed in 70% ethanol at 4° C for at least 4 h.
  • DMSO vehicle control
  • the fixed cells are centrifuged at 1500rpm for 4 minute at 4° C, washed twice with cold PBS containing 2% FBS, treated with 3 mg/ml ribonuclease (Sigma Chemical Co. Oakville, ON) and 50 ⁇ g/ml propidium iodide (PI) (Sigma Chemical Co.) for 30 minutes at 37° C.
  • the fluorescence of the stained cells is measured using a FACScan flow cytometer and the Cell Quest program (Becton Dickinson, San Jose, CA). Data are evaluated using Modfit software (Verity software House, Topsham, ME) and the effects of the selected compounds on cell cycle are evaluated.
  • DNA fragmentation analysis is used to evaluate the apoptotic effects of test compounds. Briefly, cells are plated in six-well culture plates 24 hr prior to treatment. After incubation with the test compound, medium containing detached cells is transferred to 15 ml conical tubes while cells still attached to the plate are trypsinized and then added to the same tubes. After centrifugation, collected cells are washed with PBS and resuspended in 0.5 ml lysis buffer containing 50 mM Tris-HCl, pH 8.0, 1.0 M NaCl, 10 mM EDTA and 0.5% SDS.
  • Cell lysates are transferred to microfuge tubes and proteinase K is added to a final concentration of 0.2 ml/ml and incubated overnight at 37° C.
  • DNA is extracted by phenol:chloroform:isoamyl alcohol (24:24:1), dried and dissolved in 40 ⁇ M of 10 ⁇ M Tris-HCl (pH 8.0) and 0.1 mM EDTA.
  • DNase-free RNase A is added to each sample for 30 min at 37° C and 12 ⁇ l of each sample are loaded onto a 2% agarose gel containing 0.5 ⁇ g/ml ethidium bromide and electrophoresed. DNA is visualized under UV illumination and the induction of apoptosis by the test compound is evaluated based on the generation of a nucleosomal-size DNA ladder.
  • the Matrigel Plug Assay (see, Passaniti et al., Lab. Invest. (1992) 67:519 ) is a simple method for assessing angiogenesis and the possible anti-angiogenic effect of selected compounds in mice. Briefly, liquid Matrigel (Becton Dickinson & Co., NJ) is injected subcutaneously near the abdominal midline or the dorsal flank of the animal using a 25-gauge needle.
  • Matrigel Growth factor-reduced Matrigel supplemented with 8.3 nM basic fibroblast growth factor (bFGF, Collaborative Biomedical Products, MA) stays in liquid form at 4 °C.
  • bFGF basic fibroblast growth factor
  • bFGF basic fibroblast growth factor
  • bFGF is a proven and potent inducer of angiogenesis.
  • Matrigel When injected into a mouse (0.5 ml/mouse), Matrigel immediately forms a readily recoverable solid gel, which is removed at various times (not exceeding 10 days) to assess neo-vessel growth around and into the gel. Test compounds are administered according to appropriate doses and schedules. Typically at a 5-day point, mice are sacrificed, overlying skin is removed and the gels are cut out retaining the peritoneal lining for support. For quantitation of angiogenesis, two methods are employed: 1.
  • haemoglobin content in the gel is measured using the Drabkin method ( Drabkin and Austin, J. Biol Chem. (1932) 98:719 ) and Drabkin reagent kit 525 (Sigma, MO); 2. the number of blood vessels invading the Matrigel is determined by microscopic analysis after the gels are fixed, embedded in paraffin, sectioned and stained.
  • test compounds can be evaluated in mouse xenograft models (as described above) by quantitating the effects of these compounds on tumour growth, differentiation, apoptosis and angiogenesis using immunohistochemical methods.
  • Tumour cell proliferation, angiogenesis and tumour immune infiltrates are delineated immunohistochemically using specific antibodies (Ki-67 for proliferation, CD31 for angiogenesis and NK1.1 for NK cells and F4/80 for macrophage).
  • Apoptosis is delineated utilising the TUNEL assay (In Situ Cell Death Detection kit; Boehringer Mannheim, Laval, QC). Signal generation is accomplished by peroxidase catalyzed generation of enzyme product which is visualized microscopically. Tissue histology is determined after H&E staining of separate sections.
  • tumour xenografts from treated mice are isolated, fixed and paraffin embedded individually in blocks and several 5 ⁇ m sections are cut for immunostaining and TUNEL assays. One additional section is obtained for H&E staining.
  • prior antigen retrieval is employed to improve detection.
  • a 3-step amplification method is used to generate signals in immunohistochemistry that consists essentially of applying a biotinylated secondary antibody that recognizes the primary antibody, followed by avidin-peroxidase incubation. The final step is enzyme reaction in stable DAB solution. Immunohistochemical sections are counterstained with hematoxylin for tissue histology.
  • Staining patterns are documented photographically, examined by at least two independent observers and quantitated by counting a pre-determined number of cells.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)

Description

    FIELD OF INVENTION
  • This invention pertains to the field of anti-cancer compounds and, in particular, to the therapeutically active 2,4,5-trisubstituted imidazole compounds for use in the treatment of cancer.
  • BACKGROUND OF THE INVENTION
  • A cancer is a malignant tumour of potentially unlimited growth. It is primarily the pathogenic replication (a loss of normal regulatory control) of various given types of cells found in the human body. By select mutation resulting from a primary lesion, the DNA of a cancer cell evolves and converts the cell into an autonomous system. Conventional cancer treatments have focused mainly on killing cancerous cells. Chemotherapeutic agents currently used for anti-cancer/anti-tumour therapy are selected for their toxicity towards rapidly proliferating cells. Most of them cause undesirable systemic effects such as cardiac or renal toxicity, marrow aplasia, alopecia, nausea and vomiting. During the last few years, many researchers have tried to eliminate these side effects by developing drugs having suitable physico-chemical properties allowing an increase of the availability of the drug to the tumour site. New molecules extracted from natural sources, synthetically or semi-synthetically produced, enzymes, radioisotopes, DNA toxins, various macromolecules, and antibodies against fibrin or against tumour-specific surface antigens are bound to drugs in an attempt to increase selectivity of the chemotherapeutic agents.
  • The effectiveness of most anticancer agents is greatly reduced because of their high toxicity and the nature of the illness. It is believed that the problem of high toxicity of the anticancer agents can be circumvented by chemical modifications of those structures in such a way that they act more specifically on tumour cells without increasing systemic toxicity. The research in this field is therefore mainly directed to the synthesis of anticancer agents which would possess high antineoplastic activity, low systemic toxicity and low mutagenicity on normal cells.
  • Heterocyclic compounds, especially heterocyclic azole derivatives, have been shown to have a wide spectrum of biological activities. One class of compounds with interesting biological activities is the imidazole (derivatives containing a five-membered heterocyclic azole). A variety of biological activities have been reported for imidazole derivatives with different substitution patterns (Lee et al. Nature 1994 327:739-745; Abdel-Meguid et al. Biochemistry, 1994, 33:11671; Heerding et al. Bioorg. Med. Chem. Lett. 2001, 11:2061-2065; Bu et al. Tetrahedron Lett. 1996, 37:7331-7334; Lewis JR. Nat. Prod. Rep. 1999, 16:389-418; Lewis JR. Nat. Prod. Rep. 1998, 15:417-437 and 371-395).
  • Biological activities have also been reported for aryl-imidazole derivatives, for example, these compounds can act as modulators of multi-drug resistance in cancer cells (Zhang et al. Bioorg. Med. Chem. Lett. 2000, 10:2603-2605), inhibitors of p38 MAP kinase ( Adams et al. Bioorg. Med. Chem. Lett. 2001, 11:867-2870, McLay et. al. Bioorg. Med. Chem. 2001, 9:537-554) and of cytokines ( U.S. Patent Nos. 5,656,644 ; 5,686,455 ; 5,916,891 ; 5,945,418 ; and 6,268,370 ), and inhibitors of bacterial growth ( Antolini et al. Bioorg. Med. Chem. Lett. 1999, 9:1023-1028).
  • A few reports have indicated that triaryl-imidazole compounds can act as inhibitors of p38 MAP kinase (for example, see LoGrasso et al. Biochemistry. 1997, 36:10422-10427) and as modulators of multi-drug resistance in cancer cells (Sarshar et al. Bioorg. Med. Chem. Lett. 2000,10:2599-2601), however, the majority of the literature indicates that these compounds have found use mainly as colour producing reagents. ( U.S. Patent Nos. 4,089,747 ; 5,024,935 ; 5,047,318 ; 5,496,702 ; 5,514,550 ; and 5,693,589 ) and as photopolymerization initiators ( U.S. Patent Nos. 6,117,609 and 6,060,216 ), generally in dimeric form.
  • The potential anti-cancer activity of a number of compounds has been investigated by the National Cancer Institute (NCI), which has undertaken a large scale screening of several thousand compounds to try to identify those that have potential therapeutic application in the treatment of cancer (NCI Yeast Anticancer Drug Screen). The screen is based on the ability of candidate compounds to inhibit the growth of Saccharmyces cerevisiae strains that have mutations in genes related to cell cycle control and DNA repair damage. Compounds are initially screened against a panel of six yeast strains at a single concentration (Stage0). Compounds with activity in Stage0 are re-screened against the same panel at two concentrations (Stage1). Selected compounds with activity in Stage1 that also show selectivity are re-screened against a panel of 13 yeast strains at five concentrations (Stage2). Many of the results from the screening have been made available on the NCI/DTP website. The approach adopted in this screen is dependent on a candidate compound exerting its activity on certain cellular pathways (i.e. cell cycle control or DNA repair damage). The results generated by this type of screen, therefore, represent a very preliminary stage of screening for potential anti-cancer drugs and do not necessarily correlate with the ability of a compound to inhibit the growth of cancer cells in vitro or in vivo.
  • The NCI also provides an in vivo screening program to try to identify potential anti-cancer drugs (NCI In Vivo Anticancer Drug Screen). Many of the results from this screening program are also available from the NCI/DTP website.
  • Amongst those compounds tested in one or both of the NCI screens are some aryl imidazole compounds (NCI # 322334, 338970, 144033). None of these three compounds showed any activity in the In Vivo Anticancer Drug Screen, even though one of these compounds (NCI # 338970) had been reported as active in Stage0 testing in the Yeast Anticancer Drug Screen. The fact that this compound was active in the yeast screen yet showed no activity in the in vivo assay confirms that a positive result in the yeast screen is not necessarily predictive of the utility of a compound as in anti-cancer therapeutic.
  • EP 0812829 describes tri-substituted imidazole compounds, which are useful in the treatment of cancer through sensitization of multi-drug resistant cancer cells to chemotherapeutic agents. Other compounds are described in J. Indian Chem. Soc., 1979, 56, 620-624.
  • This background information is provided for the purpose of making known information believed by the applicant to be of possible relevance to the present invention. No admission is necessarily intended, nor should be construed, that any of the preceding information constitutes prior art against the present invention.
  • SUMMARY OF THE INVENTION
  • It is an object of the present invention to provide a class of compounds which are 2,4,5-trisubstituted imidazole derivatives that have anti-cancer activity.
  • In accordance with the present invention, there is provided a compound for use in the treatment of cancer having the structural formula:
    Figure imgb0001
    or a salt thereof, wherein:
    • R4, R5, R6, R7, R8 and R9 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, or cyano or -S(O)0-2R wherein R is alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl;R10 is H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl or acyl;
    • x is CR11;
    • y is CR12 or N;
    • z is CR13 or N;
    • r is CR14 or N;
    • x' is CR15;
    • y' is CR16 or N;
    • z' is CR17 or N;
    • r' is CR18 or N;
    • R11, R12, R13, R14, R15, R16, R17 and R18 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, alkenyl, alkenyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, or cyano,
      wherein:
      • lower alkyl refers to a straight chain or branched alkyl group of 1 to 10 carbon atoms or a cyclic alkyl group of 3 to 10 carbon atoms;
      • lower alkenyl refers to a straight chain or branched hydrocarbon of 2 to 10 carbon atoms or a cyclic hydrocarbon of 3 to 10 carbon atoms, having at least one carbon to carbon double bond;
      • lower alkynyl refers to a straight chain or branched hydrocarbon of 2 to 10 carbons having at least one carbon to carbon triple bond.
  • There is also provided a compound for use in the treatment of cancer having the structural formula:
    Figure imgb0002
    or a salt thereof, wherein:
    • R4, R5, R6, R7, R8 and R9 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, cyano or -S(O)0-2R wherein R is alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl;
    • R10 is H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl or acyl;
    • R11, R12, R13, R14, R15, R16, R17 and R18 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, alkenyl, alkenyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, or cyano.
  • Also disclosed herein is a method of inhibiting neoplastic cell growth or proliferation in a mammal comprising administering to said mammal a therapeutically effective amount of a compound selected from the compounds of general formula (IV) and (VII), or a salt thereof.
  • BRIEF DESCRIPTION OF THE FIGURES
    • Figure 1 depicts the effects of a compound 92 on the proliferation of various cancer cell lines in vitro.
    • Figure 2 depicts the effects of a compound 28 on the proliferation of various cancer cell lines in vitro.
    • Figure 3 depicts the effects of a compound 50 on the proliferation of various cancer cell lines in vitro.
    • Figure 4 depicts the effects of a compound 42 on the proliferation of various cancer cell lines in vitro.
    • Figure 5A-C depicts the effects of various concentrations of a compound 45 on the proliferation of cancer cell lines in vitro at different time intervals.
    • Figure 6A-C depicts the effects of various concentrations of a compound 45 on the proliferation of cancer cell lines in vitro at different time intervals.
    • Figure 7 depicts the effects of compounds 83 and 99 on the proliferation of LS 513 colon carcinoma cells in vitro.
    • Figure 8 depicts the effects of compounds of Formula I on the proliferation of HT-29 colon adenocarcinoma cells in vitro.
    • Figure 9A-C present the cancer cell lines used to in the NCI screen used to determine the ability of compounds of Formula I to inhibit cancer cell proliferation in vitro.
    • Figure 10A depicts the average and mean GI50 values for various compounds of Formula I for a number of cancer cell lines; B depicts the average GI50 values for compound 45 by cancer cell type and C depicts the average total growth inhibition (TGI) for compound 45 by cancer cell type.
    • Figure 11 depicts the inhibition of H460 NSCLC cell proliferation in vitro by compounds of Formula I.
    • Figure 12 depicts the inhibition of HT-29 colon carcinoma cell proliferation in vitro by compounds of Formula I.
    • Figure 13 depicts the inhibition of HT-29 colon carcinoma cell proliferation in vitro by compounds of Formula I.
    • Figure 14 depicts the effects of compounds of Formula I on the growth of HT-29 colon adenocarcinoma cells in vivo in CD-1 nude mice.
    • Figure 15 depicts the effects of compounds of Formula I on the average weight of tumours in CD-1 nude mice (average weight per group of mice).
    • Figure 16 depicts the effects of compounds of Formula I on the weight of tumours in CD-1 nude mice (individual tumour weights).
    • Figure 17 depicts the effects of compounds of Formula I on the growth of HT-29 colon adenocarcinoma cells in vivo in CD-1 nude mice.
    • Figure 18 depicts the effect of compound 45 on the growth of HepG2 hepatocarcinoma cells in vivo in CD-1 nude mice in terms of A tumour size, and B tumour weight.
    • Figure 19 depicts the effects of compounds 45, 33 and 90 on the activity of various human kinases.
    • Figure 20 depicts the subcellular location of compound 45 in HT-29 colon adenocarcinoma cells (A, B); of doxorubicin in HT-29 colon adenocarcinoma cells (C); of compound 45 in A498 renal cancer cells (D), and of compound 45 in C8161 melanoma cells (E).
    • Figure 21 depicts the formation of vacuoles in HT-29 colon adenocarcinoma cells treated with compound 45 or doxorubicin.
    • Figure 22 depicts the effects of compound 45 on the cell cycle in HT-29 colon adenocarcinoma cells.
    DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a class of 2,4,5-trisubstituted imidazole compounds and them for use as anti-cancer agents.
  • Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains.
  • The terms are defined as follows:
    • The term "halogen" refers to fluorine, bromine, chlorine, and iodine atoms.
    • The term "hydroxyl" refers to the group -OH.
    • The term "thiol" or "mercapto" refers to the group -SH, and -S(O)0-2.
    • The term "lower alkyl" refers to a straight chain or branched alkyl group of one to ten carbon atoms or a cyclic alkyl group of three to ten carbon atoms. This term is further exemplified by such groups as methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, 1-butyl (or 2-methylpropyl), cyclopropylmethyl, i-amyl, n-amyl, hexyl and the like.
  • The term "substituted lower alkyl" refers to lower alkyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom of the lower alkyl moiety.
  • The term "lower alkenyl" refers to a straight chain or branched hydrocarbon of two to ten carbon atoms or a cyclic hydrocarbon of three to ten carbon atoms, having at least one carbon to carbon double bond.
  • The term "substituted lower alkenyl" refers to lower alkenyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl, alkenyl, alkynyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom to produce a stable compound.
  • The term "lower alkynyl" refers to a straight chain or branched hydrocarbon of two to ten carbon atoms having at least one carbon to carbon triple bond.
  • The term "substituted lower alkynyl" refers to lower alkynyl as just described including one or more groups such as hydroxyl, thiol, alkylthiol, halogen, alkoxy, amino, amido, carboxyl, cycloalkyl, substituted cycloalkyl, heterocycle, cycloheteroalkyl, substituted cycloheteroalkyl, acyl, carboxyl, aryl, substituted aryl, aryloxy, hetaryl, substituted hetaryl, aralkyl, heteroaralkyl, alkyl, alkenyl, alkynyl, alkyl alkenyl, alkyl alkynyl, alkyl cycloalkyl, alkyl cycloheteroalkyl, nitro, cyano. These groups may be attached to any carbon atom to produce a stable compound.
  • The term "alkoxy" refers to the group -OR, where R is lower alkyl, substituted lower alkyl, acyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heteroalkyl, heteroarylalkyl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, or substituted cycloheteroalkyl as defined below.
  • The term "alkylthio" denotes the group -SR, -S(O)n=1-2 -R, where R is lower alkyl, substituted lower alkyl, aryl, substituted aryl, aralkyl or substituted aralkyl as defined below.
  • The term "acyl" refers to groups -C(O)R, where R is hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, cycloalkyl or substituted cycloalkyl.
  • The term "aryloxy" refers to groups -OAr, where Ar is an aryl, substituted aryl, heteroaryl, or substituted heteroaryl group as defined below.
  • The term "amino" refers to the group NRR', where R and R' may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, heteroaryl, cycloalkyl, or substituted heteroaryl as defined below, acyl, D or L aminoacid or a protected form thereof.
  • The term "amido" refers to the group -C(O)NRR', where R and R' may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, hetaryl, substituted hetaryl as defined below.
  • The term "carboxyl" refers to the group -C(O)OR, where R may independently be hydrogen, lower alkyl, substituted lower alkyl, aryl, substituted aryl, hetaryl, substituted hetaryl and the like as defined.
  • The terms "aryl" or "Ar" refer to an aromatic carbocyclic group having at least one aromatic ring (e.g., phenyl or biphenyl) or multiple condensed rings in which at least one ring is aromatic, (e.g., 1,2,3,4-tetrahydronaphthyl, naphthyl, anthryl, phenanthryl, 9-fluorenyl, dibenzocycloheptatrienyl etc.).
  • The term "substituted aryl" refers to aryl optionally substituted with one or more functional groups, e.g., halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl, substituted heteroaryl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido, cyano or N=CRR', wherein R and R' are independemtly selected from H, alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl or substituted heteroaryl.
  • The term "heterocycle" refers to a saturated, unsaturated, or aromatic carbocyclic group having a single ring (e.g., morpholino, pyridyl or furyl) or multiple condensed rings (e.g., naphthpyridyl, quinoxalyl, quinolinyl, indolizinyl, indanyl or benzo[b]thienyl) and having at least one hetero atom, such as N, O or S, within the ring.
  • The term "substituted heterocycle" refers to heterocycle optionally substituted with, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl, substituted heteroaryl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido or cyano and the like.
  • The terms "heteroaryl" or "hetaryl" refer to a heterocycle in which at least one heterocyclic ring is aromatic.
  • The term "substituted heteroaryl" refers to a heterocycle optionally mono or poly substituted with one or more functional groups, e.g., halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl, substituted heteroaryl, heteroalkyl, substituted heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido, cyano or -N=CRR', wherein R and R' are independemtly selected from H, alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, substituted heterocycle, heteroaryl or substituted heteroaryl and the like.
  • The term "aralkyl" refers to the group -R-Ar where Ar is an aryl group and R is lower alkyl or substituted lower alkyl group. Aryl groups can optionally be unsubstituted or substituted with, e.g., halogen, lower alkyl, alkoxy, alkyl thio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • The term "heteroalkyl" refers to the group -R-Het where Het is a heterocycle group and R is a lower alkyl group. Heteroalkyl groups can optionally be unsubstituted or substituted with e.g., halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • The term "heteroarylalkyl" refers to the group -R-HetAr where HetAr is an heteroaryl group and R lower alkyl or substituted loweralkyl. Heteroarylalkyl groups can optionally be unsubstituted or substituted with, e.g., halogen, lower alkyl, substituted lower alkyl, alkoxy, alkylthio, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • The term "cycloalkyl" refers to a cyclic or polycyclic alkyl group containing 3 to 15 carbon. For polycyclic groups, these may be multiple condensed rings in which one of the distal rings maybe aromatic (e.g. tetrahydronaphthalene, etc.)..
  • The term "substituted cycloalkyl" refers to a cycloalkyl group comprising one or more substituents with, e.g halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, trifluoromethyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, sulfamido or cyano and the like.
  • The term "cycloheteroalkyl" refers to a cycloalkyl group wherein one or more of the ring carbon atoms is replaced with a heteroatom (e.g., N, O, S or P).
  • The term "substituted cycloheteroalkyl" refers to a cycloheteroalkyl group as herein defined which contains one or more substituents, such as halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • The term "alkyl cycloalkyl" refers to the group -R-cycloalkyl where cycloalkyl is a cycloalkyl group and R is a lower alkyl or substituted lower alkyl. Cycloalkyl groups can optionally be unsubstituted or substituted with e.g. halogen, lower alkyl, lower alkoxy, lower alkylthio, trifluoromethyl, amino, amido, carboxyl, hydroxyl, aryl, aryloxy, heterocycle, hetaryl, substituted hetaryl, nitro, cyano, alkylthio, thiol, sulfamido and the like.
  • The terms "therapy" and "treatment," as used interchangeably herein, refer to an intervention performed with the intention of alleviating the symptoms associated with, preventing the development of, or altering the pathology of a disease, disorder or condition. Thus, the terms therapy and treatment are used in the broadest sense, and include the prevention (prophylaxis), moderation, reduction, and curing of a disease, disorder or condition at various stages. Those in need of therapy/treatment include those already having the disease, disorder or condition as well as those prone to, or at risk of developing, the disease, disorder or condition and those in whom the disease, disorder or condition is to be prevented.
  • The term "subject" or "patient," as used herein, refers to an animal in need of treatment.
  • The term "animal," as used herein, refers to both human and non-human animals, including, but not limited to, mammals, birds and fish.
  • Administration of the compounds of the invention "in combination with" one or more further therapeutic agents, is intended to include simultaneous (concurrent) administration and consecutive administration. Consecutive administration is intended to encompass various orders of administration of the therapeutic agent(s) and the compound(s) of the invention to the subject.
  • As used herein, the term "about" refers to a +/-10% variation from the nominal value. It is to be understood that such a variation is always included in any given value provided herein, whether or not it is specifically referred to.
  • I . 2,4,5-Trisubstituted Imidazole Compounds
  • The present invention includes the compound of the structural formula, for use in the treatment of cancer:
    Figure imgb0003
    or a salt thereof, wherein:
    • R4, R5, R6, R7, R8 and R9 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, cyano or -S(O)0-2R wherein R is alkyl,
    • substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl;
    • x is CR11;
    • y is CR12 or N;
    • z is CR13 or N;
    • r is CR1 or N;
    • x' is CR15;
    • y' is CR16 or N;
    • z' is CR17 or N;
    • r' is CR18 or N;
    • R10 is H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl or acyl; and
    • R11, R12, R13, R14, R15, R16, R17 and R18 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, or cyano.
  • There is also provided the compound of the compound of the structural formula, for use in the treatment of cancer:
    Figure imgb0004
    or a salt thereof, wherein:
    • R4, R5, R6, R7, R8 and R9 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, or cyano or -S(O)0-2R wherein R is alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl;
    • R10 is H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl or acyl;
    • R11, R12, R13, R14, R15, R16, R17 and R18 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro or cyano. Compounds of the present invention include the following exemplary compounds:
      Figure imgb0005
      Figure imgb0006
      Figure imgb0007
      Figure imgb0008
      Figure imgb0009
      Figure imgb0010
      Figure imgb0011
      Figure imgb0012
      Figure imgb0013
      Figure imgb0014
      Figure imgb0015
      Figure imgb0016
      Figure imgb0017
      Figure imgb0018
      Figure imgb0019
      Figure imgb0020
      Figure imgb0021
      Figure imgb0022
      Figure imgb0023
      Figure imgb0024
      Figure imgb0025
      Figure imgb0026
      Figure imgb0027
      Figure imgb0028
      Figure imgb0029
      Figure imgb0030
      Figure imgb0031
      Figure imgb0032
      Figure imgb0033
      Figure imgb0034
      Figure imgb0035
      Figure imgb0036
      Figure imgb0037
      Figure imgb0038
      Figure imgb0039
      Figure imgb0040
      Figure imgb0041
      Figure imgb0042
      Figure imgb0043
      Figure imgb0044
      Figure imgb0045
      Figure imgb0046
      Figure imgb0047
      Figure imgb0048
  • The present invention includes pharmaceutically acceptable salts of the compounds defined by Formula VI. Compounds according to the present invention can possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with a number of organic and inorganic bases, and organic and inorganic acids, to form pharmaceutically acceptable salts.
  • The term "pharmaceutically acceptable salt" as used herein, refers to a salt of a compound of Formula VI, which is substantially non-toxic to living organisms. Typical pharmaceutically acceptable salts include those salts prepared by reaction of the compound of the present invention with a pharmaceutically acceptable mineral or organic acid or an organic or inorganic base. Such salts are known as acid addition and base addition salts.
  • Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulphonic acid, methanesulphonic acid, oxalic acid, p-bromophenylsulphonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like. Examples of such pharmaceutically acceptable salts are the sulphate, pyrosulphate, bisulphate, sulphite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, hydrochloride, dihydrochloride, isobutyrate, caproate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1,4-dioate, hexyne-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, hydroxybenzoate, methoxybenzoate, phthalate, xylenesulphonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, gamma-hydroxybutyrate, glycolate, tartrate, methanesulphonate, propanesulphonate, naphthalene-1-sulfonate, napththalene-2-sulfonate, mandelate and the like. Preferred pharmaceutically acceptable acid addition salts are those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and those formed with organic acids such as maleic acid and methanesulphonic acid.
  • Salts of amine groups may also comprise quarternary ammonium salts in which the amino nitrogen carries a suitable organic group such as an alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, or aralkyl moiety.
  • Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like. Bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, calcium hydroxide, calcium carbonate, and the like.
  • One skilled in the art will understand that the particular counterion forming a part of a salt of this invention is usually not of a critical nature, so long as the salt as a whole is pharmacologically acceptable and as long as the counterion does not contribute undesired qualities to the salt as a whole. Also disclosed herein are pharmaceutically acceptable solvates of a compound of Formula VI. Many of the compounds of Formula VI can combine with solvents such as water, methanol, ethanol
    and acetonitrile to form pharmaceutically acceptable solvates such as the corresponding hydrate, methanolate, ethanolate and acetonitrilate.
  • The compounds of the present invention may have multiple asymmetric (chiral) centres. As a consequence of these chiral centres, the compounds of the present invention occur as racemates, mixtures of enantiomers and as individual enantiomers, as well as diastereomers and mixtures of diastereomers. All asymmetric forms, individual isomers and combinations thereof, are within the scope of the present invention.
  • It will be readily understood by one skilled in the art that if the stereochemistry of a compound of Formula I is critical to its activity, then the relative stereochemistry of the compound is established early during synthesis to avoid subsequent stereoisomer separation problems. Further manipulation of the molecule will then employ stereospecific procedures so as to maintain the desired chirality.
  • Non-toxic metabolically-labile esters or amides of a compound of Formula VI are those that are hydrolysed in vivo to afford the compound of Formula VI and a pharmaceutically acceptable alcohol or amine. Examples of metabolically-labile esters include esters formed with (1-6C) alkanols, in which the alkanol moiety may be optionally substituted by a (1-8C) alkoxy group, for example methanol, ethanol, propanol and methoxyethanol. Non-limiting examples of metabolically-labile amides include amides formed with amines such as methylamine.
  • II . Preparation of Compounds
  • As is known in the art, triaryl imidazole compounds can be prepared by a number of standard techniques. Compounds, therefore, can be prepared by several general synthetic methods, for example, as described by Grimmett, (Grimmett, M.R., Comprehensive Heterocyclic Chemistry: The Structure, Reaction, Synthesis and Uses of Heterocyclic Compounds, A. R. Katrizky and C. W. Rees, eds., Vol. 5, Pergamon Press. Oxford, 1984, pp. 457-498; Grimmett, M. R., Imidazole and Benzimidazole Synthesis, Academic Press, San Diego CA, 1997).
  • Compounds are prepared via solution or solid phase synthesis, by reacting a dione with the aldehyde at elevated temperature in the presence of ammonium acetate in acetic acid (see, for example, Krieg et al., Naturforsch. 1967, 22b:132; Sarshar et al., Tetrahedron Lett. 1996, 37:835-838).
    Figure imgb0049
  • The compounds of Formula (XXXI) and (XXXII) are either commercially available or may be prepared using standard procedures known to a person skilled in the relevant art. Compounds of Formula (XXXI), therefore, can be prepared by several general synthetic methods, for example, as described by: Fischer et. al (J. Am. Chem. Soc. 1961, 83, 4208-4210); Guijarro et al. (J. Am. Chem. Soc. 1999, 121, 4155-4157); Chi et. al. (Synth. Comm. 1994, 24(15), 2119-2122) and Armesto et. al. (Synthesis, 1988, 799-801).
  • Compounds of formula XXXI can also be prepared:
    1. i) by oxidizing a compound of formula (XXXIII). Compounds of formula (XXXIII), in turn can be prepared by reacting a compounds of formula (XXXIV) with sodium cyanide in the presence of a solvent as shown below, wherein R3 = R2 and R2 is as defined above:
      Figure imgb0050
      or,
    2. ii) by oxidizing a compound of formula (XXXV). Compounds of formula (XXXV), in turn can be prepared by treating a compound of formula (XXXIV) and a compound of formula (XXXVI) with sodium cyanide in the presence of a solvent as shown below, wherein R2 and R3 are as defined above:
      Figure imgb0051
      or,
    3. iii) by oxidizing a compound of formula (XXXVII). Compounds of formula (XXXVII) in turn can be prepared by oxidizing a compound of formula (XXVIII) or (XXXIX) as shown below, wherein R2 and R3 are as defined above:
      Figure imgb0052
      or,
    4. iv) by oxidizing a compound of formula (XXXIX) using PdCl2 in DMSO, or,
    5. v) by deprotecting and oxidizing a compound of formula (XL). Compounds of formula (XL) in turn can be prepared by reacting a compound of formula (XLI) with a compound of formula (XLII) in the presence of a suitable base:
      Figure imgb0053
      wherein R2 and R3 are independently, aryl, substituted aryl, heteroaryl or substituted heteroaryl,
      or,
    6. vi) by reacting a compound of formula (XLIII) with a substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl under Friedel-Crafts acylation conditions or by nucleophilic displacement of the chloride in compound of formula (XLIII). Compounds of formula (XLIII) in turn can be prepared by reacting a substituted or unsubstituted aryl or substituted heteroaryl or unsubstituted heteroaryl with oxalyl chloride under Friedel-Crafts acylation conditions:
      Figure imgb0054
      wherein R2 and R3 are independently aryl, substituted aryl, heteroaryl or substituted heteroaryl;
      or
    7. vii) by oxidising a compound of formula (XLIV). Compounds of formula (XLIV) in turn can be prepared by reacting a compound of formula (XLV) with thionyl chloride in benzene with catalytic dimethylformamide to form an intermediate (XLVI). This intermediate (XLVI) is then used directly without purification in a Freidel-Crafts reaction to produce the ketone (XLIV).
      Figure imgb0055
    III. Anti-cancer Activity of Compounds of the invention
  • The ability of a candidate compound of Formula VI to inhibit neoplastic cell growth and/or proliferation can be tested using standard techniques known in the art. In addition, compounds of Formula VI that demonstrate inhibitory activity may be further tested in vitro and/or in vivo in combination with various known chemotherapeutics to evaluate their potential use in combination therapies. Exemplary methods of testing candidate compounds are provided below and in the Examples included herein. One skilled in the art will understand that other methods of testing the compounds are known in the art and are also suitable for testing candidate compounds.
  • A. In vitro Testing
  • Candidate compounds of the invention can be assayed initially in vitro for their ability to inhibit cell growth (i.e. their cytotoxicity) using standard techniques. In general, cells of a specific test cell line (typically a cancer cell line) are grown to a suitable density (e.g. approximately 1 x 104) and the candidate compound is added. After an appropriate incubation time (typically between about 48 to 74 hours), cell survival is assessed, for example, by assaying for tetrazolium salt (or modified tetrazolium salt) cleavage, or by using the resazurin reduction test (see Fields & Lancaster (1993) Am. Biotechnol. Lab. 11:48-50; O'Brien et al., (2000) Eur J. Biochem. 267:5421-5426 and U.S. Patent No. 5,501,959 ), the sulforhodamine assay (Rubinstein et al., (1990) J. Natl. Cancer Inst. 82:113-118) or the neutral red dye test (Kitano et al., (1991) Euro. J. Clin. Investg. 21:53-58; West et al., (1992) J. Investigative Derm. 99:95-100). Inhibition of cell growth is determined by comparison of cell survival in the treated culture with cell survival in one or more control cultures, for example, cultures not pre-treated with the candidate compound and/or those pre-treated with a control compound (typically a known therapeutic). Other suitable techniques for assessing cytotoxicity are known in the art.
  • Assays that measure metabolic activity (such as tetrazolium-based assays) can also be used to assess the effect of candidate compounds on cell activation and /or proliferation, due the fact that proliferating cells are metabolically more active than resting cells.
  • Candidate compounds can also be tested in vitro for their ability to inhibit anchorage-independent growth of tumour cells. Anchorage-independent growth is known in the art to be a good indicator of tumourigenicity. In general, anchorage-independent growth is assessed by plating cells from an appropriate cancer cell-line onto soft agar and determining the number of colonies formed after an appropriate incubation period. Growth of cells treated with the candidate compound can then be compared with that of cells treated with an appropriate control (as described above).
  • A variety of cancer cell-lines suitable for testing the candidate compounds are known in the art. In one embodiment, in vitro testing of the candidate compounds is conducted in a human cancer cell-line. Examples of suitable human cancer cell-lines for in vitro testing of the compounds of the present invention include, but are not limited to, colon and colorectal carcinoma cell lines such as HT-29, CaCo, LoVo, COLO320 and HCT-116; non small cell lung cancer cell lines such as NCI-H460, small cell lung cancer cell lines such as H209; breast cancer cell lines such as MCF-7, T47D and MDA-MB-231; ovarian cancer cell lines such as SK-OV-3; prostate cancer cell lines such as PC-3 and DU-145; chronic myeloid leukemia cell lines such as K562; bladder cancer cell lines such as T24; brain cancer cell lines such as U-87-MG; pancreatic cancer cell lines such as AsPC-1, SU.86.86 and BxPC-3; kidney cancer cell lines such as A498 and Caki-1; liver cancer cell lines such as HepG2, and skin cancer cell lines such as A2058 and C8161. Drug-resistant cancer cell lines can be used to determine the ability of the compounds of the present invention to inhibit growth and/or proliferation of drug- or multi-drug resistant neoplastic cells.
  • The selectivity of the candidate compounds of the invention may also be tested, i.e. the ability of the compound to demonstrate some level of selective action toward neoplastic (or cancer) cells in comparison to normal proliferating cells. An exemplary method of assessing the differential sensitivity between normal and cancer cells for a compound has been described by Vassilev et al. (Anti-Cancer Drug Design (2001) 16:7). This method involves the comparison of IC90 values, i.e. the molar concentration of a test compound required to cause 90% growth inhibition of exponentially growing cells. Thus, the IC90 values for candidate compounds can be evaluated in various cancer cell lines (such as those outlined above) and normal cells (such as HUVEC and/or WI38 cells) and compared. IC90 values can be measured using a variety of standard techniques including those described above for cytotoxicity testing.
  • While the mechanism of action of the compounds of the inventionis not relevant to the instant invention, assays to investigate potential mechanisms of action of the compounds may be conducted if desired in order to provide information useful in determining what aspects of tumour growth the compounds affect. This type of information may help to determine cancer types that will benefit from treatment with the compounds. Examples of such assays include, but are not limited to, cell-cycle analysis (for example, employing flow cytometry techniques), apoptosis assays (such as DNA fragmentation analysis), anti-angiogenesis assays (for example, various Matrigel assays, including cord formation and Matrigel plug assays) and immunohistochemical analysis.
  • Toxicity of the candidate compounds can also be initially assessed in vitro using standard techniques. For example, human primary fibroblasts can be treated in vitro with a compound of the invention and then tested at different time points following treatment for their viability using a standard viability assay, such as the assays described above or the trypan-blue exclusion assay. Cells can also be assayed for their ability to synthesize DNA, for example, using a thymidine incorporation assay, and for changes in cell cycle dynamics, for example, using a standard cell sorting assay in conjunction with a fluorocytometer cell sorter (FACS).
  • B. In vivo Testing
  • The ability of the candidate compounds to inhibit tumour growth, proliferation and/or metastasis in vivo can be determined in an appropriate animal model using standard techniques known in the art (see, for example, Enna, et al., Current Protocols in Pharmacology, J. Wiley & Sons, Inc., New York, NY). Exemplary protocols are provided below and in the Examples. Non-limiting examples of suitable animal models are provided in Table 1.
  • In general, current animal models for screening anti-tumour compounds are xenograft models, in which a human tumour has been implanted into an animal. For example, the candidate compounds can be tested in vivo on solid tumours using mice that are subcutaneously grafted or injected with 30 to 60 mg of a tumour fragment, or an appropriate number of tumour cells (e.g. about 106 to 107) on day 0. The animals bearing tumours are mixed before being subjected to the various treatments and controls. In the case of treatment of advanced tumours, tumours are allowed to: develop to the desired size, animals having insufficiently developed tumours being eliminated. The selected animals are distributed at random to undergo the treatments and controls. Animals not bearing tumours may also be subjected to the same treatments as the tumour-bearing animals in order to be able to dissociate the toxic effect from the specific effect on the tumour. Chemotherapy generally begins from 3 to 22 days after grafting, depending on the type of tumour, and the animals are observed every day. Candidate compounds can be administered to the animals, for example, by bolus infusion. The different animal groups are weighed about 3 or 4 times a week until the maximum weight loss is attained, after which the groups are weighed at least once a week until the end of the trial.
  • The tumours are measured about 2 or 3 times a week until the tumour reaches a pre-determined size and / or weight, or until a pre-determined time period has passed, or until the animal dies (if this occurs before the tumour reaches the pre-determined size / weight). The animals are then sacrificed and the tissue histology, size and / or proliferation of the tumour assessed.
  • If desired, one or more standard immunohistochemical tests may also be conducted on tissues isolated from the test animals in order to determine the effects of the compound on tumour growth, differentiation, apoptosis and/or angiogenesis. Examples of such tests include, but are not limited to, the use of specific antibodies (for example, antibodies against Ki-67 to assess proliferation, CD31 to assess angiogenesis, NK1.1 as an indication of the presence of NK cells, F4/80 as an indication of the presence of macrophages) and TUNEL assays to determine apoptosis.
  • Other models, such as orthopedic implantation of tumours into animals (i.e. the implantation of cancer cells of a certain type into the corresponding tissue in the animal, such as pancreatic cancer cells into the pancreas), may also be used to assess the effect of the candidate compounds on tumour growth and proliferation. In addition, the effect of the candidate compound on spontaneous tumours in normal mice can be assessed.
  • The effect of the candidate compounds on drug-resistant tumours can be assessed in vivo by utilising a drug- or multidrug-resistant cancer cell in the xenograft experiments.
  • For the study of the effect of the candidate compounds on haematologic tumours, such as lymphomas or leukaemias, the animals are grafted or injected with a particular number of cells, and the anti-tumour activity is determined by the increase in the survival time of the treated mice relative to the controls.
  • To study the effect of the candidate compounds on tumour metastasis, tumour cells are typically treated with the compound ex vivo and then injected into a suitable test animal. The spread of the tumour cells from the site of injection is then monitored over a suitable period of time.
  • The ability of the candidate compounds to act in combination with, or to sensitise a tumour to the effects of, another chemotherapeutic agent can also be tested in the above models. In this case, the test animals would be treated with both the chemotherapeutic agent and the candidate compound of the invention. Control animal could include animals treated with the chemotherapeutic alone, animals treated with the candidate compound alone and/or untreated animals.
  • In vivo toxic effects of the compounds of the invention can be evaluated by standard techniques, for example, by measuring their effect on animal body weight during treatment and by performing haematological profiles and liver enzyme analysis after the animal has been sacrificed (survival assays). Table I: Examples of in vivo models of human cancer
    Cancer Model Cell Type
    Tumour Growth Assay Prostate (PC-3, DU145)
    Human solid tumour xenografts in mice (sub-cutaneous injection) Breast (MDA-MB-231, MVB-9)
    Colon (HT-29)
    Lung (NCI-H460, NCI-H209)
    Pancreatic (ASPC-1, SU86.86)
    Pancreatic: drug resistant (BxPC-3)
    Skin (A2058, C8161)
    Cervical (SIHA, HeLa-S3)
    Cervical: drug resistant (HeLa S3-
    HU-resistance)
    Liver (HepG2)
    Brain (U87-MG)
    Renal (Caki-1, A498)
    Ovary (SK-OV-3)
    Bladder (T24)
    Tumour Growth Assay Breast: drug resistant (MDA-CDDP-S4, MDA-MB435-To.1)
    Human solid tumour isografts in mice (fat pad injection)
    Survival Assay Human: Burkitts lymphoma (Non-Hodgkin's) (raji)
    Experimental model of lymphoma and leukaemia in mice Murine: erythroleukemia (CB7 Friend retrovirus-induced)
    Experimental model of lung metastasis in mice Human: melanoma (C8161)
    Murine: fibrosarcoma (R3)
  • IV. Toxicity Testing
  • It is important that the anti-cancer compounds of the present invention exhibit low toxicity in vivo. Toxicity tests for potential drugs are well-known in the art (see, for example, Hayes, A.W., ed., (1994), Principles and Methods of Toxicology, 3rd ed., Raven Press, NY; Maines, M., ed., Current Protocols in Toxicology, John Wiley & Sons, Inc., NY).
  • In vitro acute toxicity testing of a compound of the invention can be performed using mammalian cell lines (see, for example, Ekwall, B., Ann. N.Y. Acad. Sci., (1983) 407:64-77). Selection of an appropriate cell line is dependent on the potential application of the candidate compound and can be readily determined by one skilled in the art.
  • In vivo toxicity testing can be performed by standard methodology. For example, by injecting varying concentrations of the candidate compound into an appropriate animal model. The compound can be injected once, or administration can be repeated over several days. The toxic effects of the compound can be evaluated over an appropriate time period by monitoring the general health and body weight of the animals. After the completion of the period of assessment, the animals can be sacrificed and the appearance and weight of the relevant organs determined. An indication of the toxicity of a compound can also be obtained during the in vivo anti-cancer testing of the compound.
  • V. Therapeutic Uses of Compounds of the invention
  • The compounds of the invention can be used in the treatment and/or stabilisation of various types of cancers. In this context, the compounds may exert either a cytotoxic or cytostatic effect resulting in a reduction in the size of a tumour, the slowing or prevention of an increase in the size of a tumour, an increase in the disease-free survival time between the disappearance or removal of a tumour and its reappearance, prevention of an initial or subsequent occurrence of a tumour (e.g. metastasis), an increase in the time to progression, reduction of one or more adverse symptom associated with a tumour, or an increase in the overall survival time of a subject having cancer. The compounds can be used alone or they can be used as part of a multi-drug regimen in combination with one or more known therapeutics.
  • Examples of cancers which may be may be treated or stabilized in accordance with the present invention include, but are not limited to haematologic neoplasms, including leukaemias and lymphomas; carcinomas, including adenocarcinomas; melanomas and sarcomas. Carcinomas, adenocarcinomas and sarcomas are also frequently referred to as "solid tumours." Examples of commonly occurring solid tumours include, but are not limited to, cancer of the brain, breast, cervix, colon, head and neck, kidney, lung, ovary, pancreas, prostate, stomach and uterus, non-small cell lung cancer and colorectal cancer. Various forms of lymphoma also may result in the formation of a solid tumour and, therefore, are also often considered to be solid tumours. One embodiment of the present invention provides for the use of the compounds of Formula VI in the treatment and/or stabilisation of a solid tumour.
  • The term "leukaemia" refers broadly to progressive, malignant diseases of the blood-forming organs. Leukaemia is typically characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow but can also refer to malignant diseases of other blood cells such as erythroleukaemia, which affects immature red blood cells. Leukaemia is generally clinically classified on the basis of (1) the duration and character of the disease - acute or chronic; (2) the type of cell involved - myeloid (myelogenous), lymphoid (lymphogenous) or monocytic, and (3) the increase or non-increase in the number of abnormal cells in the blood - leukaemic or aleukaemic (subleukaemic). Leukaemia includes, for example, acute nonlymphocytic leukaemia, chronic lymphocytic leukaemia, acute granulocytic leukaemia, chronic granulocytic leukaemia, acute promyelocytic leukaemia, adult T-cell leukaemia, aleukaemic leukaemia, aleukocythemic leukaemia, basophylic leukaemia, blast cell leukaemia, bovine leukaemia, chronic myelocytic leukaemia, leukaemia cutis, embryonal leukaemia, eosinophilic leukaemia, Gross' leukaemia, hairy-cell leukaemia, hemoblastic leukaemia, hemocytoblastic leukaemia, histiocytic leukaemia, stem cell leukaemia, acute monocytic leukaemia, leukopenic leukaemia, lymphatic leukaemia, lymphoblastic leukaemia, lymphocytic leukaemia, lymphogenous leukaemia, lymphoid leukaemia, lymphosarcoma cell leukaemia, mast cell leukaemia, megakaryocytic leukaemia, micromyeloblastic leukaemia, monocytic leukaemia, myeloblastic leukaemia, myelocytic leukaemia, myeloid granulocytic leukaemia, myelomonocytic leukaemia, Naegeli leukaemia, plasma cell leukaemia, plasmacytic leukaemia, promyelocytic leukaemia, Rieder cell leukaemia, Schilling's leukaemia, stem cell leukaemia, subleukaemic leukaemia, and undifferentiated cell leukaemia.
  • The term "sarcoma" generally refers to a tumour which originates in connective tissue, such as muscle, bone, cartilage or fat, and is made up of a substance like embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance. Sarcomas include soft tissue sarcomas, chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abemethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumour sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, granulocytic sarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented haemorrhagic sarcoma, immunoblastic sarcoma of B cells, lymphoma, immunoblastic sarcoma of T-cells, Jensen's sarcoma, Kaposi's sarcoma, Kupffer cell sarcoma, angiosarcoma, leukosarcoma, malignant mesenchymoma sarcoma, parosteal sarcoma, reticulocytic sarcoma, Rous sarcoma, serocystic sarcoma, synovial sarcoma, and telangiectaltic sarcoma.
  • The term "melanoma" is taken to mean a tumour arising from the melanocytic system of the skin and other organs. Melanomas include, for example, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma, Harding-Passey melanoma, juvenile melanoma, lentigo maligna melanoma, malignant melanoma, nodular melanoma, subungal melanoma, and superficial spreading melanoma.
  • The term "carcinoma" refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases. Exemplary carcinomas include, for example, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colorectal carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiermoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex ulcere, carcinoma fibrosum, gelatiniform carcinoma, gelatinous carcinoma, giant cell carcinoma, carcinoma gigantocellulare, glandular carcinoma, granulosa cell carcinoma, hair-matrix carcinoma, haematoid carcinoma, hepatocellular carcinoma, Hurthle cell carcinoma, hyaline carcinoma, hypemephroid carcinoma, infantile embryonal carcinoma, carcinoma in situ, intraepidermal carcinoma, intraepithelial carcinoma, Krompecher's carcinoma, Kulchitzky-cell carcinoma, large-cell carcinoma, lenticular carcinoma, carcinoma lenticulare, lipomatous carcinoma, lymphoepithelial carcinoma, carcinoma medullare, medullary carcinoma, melanotic carcinoma, carcinoma molle, mucinous carcinoma, carcinoma muciparum, carcinoma mucocellulare, mucoepidermoid carcinoma, carcinoma mucosum, mucous carcinoma, carcinoma myxomatodes, naspharyngeal carcinoma, oat cell carcinoma, non-small cell carcinoma, carcinoma ossificans, osteoid carcinoma, papillary carcinoma, periportal carcinoma, preinvasive carcinoma, prickle cell carcinoma, pultaceous carcinoma, renal cell carcinoma of kidney, reserve cell carcinoma, carcinoma sarcomatodes, schneiderian carcinoma, scirrhous carcinoma, carcinoma scroti, signet-ring cell carcinoma, carcinoma simplex, small-cell carcinoma, solanoid carcinoma, spheroidal cell carcinoma, spindle cell carcinoma, carcinoma spongiosum, squamous carcinoma, squamous cell carcinoma, string carcinoma, carcinoma telangiectaticum, carcinoma telangiectodes, transitional cell carcinoma, carcinoma tuberosum, tuberous carcinoma, verrucous carcinoma, and carcinoma villosum.
  • The term "carcinoma" also encompasses adenocarcinomas. Adenocarcinomas are carcinomas that originate in cells that make organs which have glandular (secretory) properties or that originate in cells that line hollow viscera, such as the gastrointestinal tract or bronchial epithelia. Examples include, but are not limited to, adenocarcinomas of the breast, lung, colon, pancreas and prostate.
  • Additional cancers encompassed by the present invention include, for example, Hodgkin's Disease, Non-Hodgkin's lymphoma, multiple myeloma, neuroblastoma, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small-cell lung tumours, primary brain tumours, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, gliomas, testicular cancer, thyroid cancer, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, mesothelioma and medulloblastoma.
  • The cancer to be treated or stabilized may be indolent or it may be aggressive. The compounds of the invention can be used to treat refractory cancers, advanced cancers, recurrent cancers and metastatic cancers. One skilled in the art will appreciate that many of these categories may overlap, for example, aggressive cancers are typically also metastatic.
  • "Aggressive cancer," as used herein, refers to a rapidly growing cancer. One skilled in the art will appreciate that for some cancers, such as breast cancer or prostate cancer the term "aggressive cancer" will refer to an advanced cancer that has relapsed within approximately the earlier two-thirds of the spectrum of relapse times for a given cancer, whereas for other types of cancer, such as small cell lung carcinoma (SCLC), nearly all cases present rapidly growing cancers which are considered to be aggressive. The term can thus cover a subsection of a certain cancer type or it may encompass all of other cancer types. A "refractory" cancer or tumour refers to a cancer or tumour that has not responded to treatment. "Advanced cancer," refers to overt disease in a patient that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy. Advanced disease may refer to a locally advanced cancer or it may refer to metastatic cancer. The term "metastatic cancer" refers to cancer that has spread from one part of the body to another.
  • Also disclose is the use of the compounds of Formula VI as "sensitizing agents," which selectively inhibit the growth of cancer cells. In this case, the compound alone does not have a cytotoxic effect on the cancer cell, but provides a means of weakening the cancer cells, and thereby facilitates the benefit from conventional anti-cancer therapeutics.
  • Thus, disclosed herein is the administration to a subject of a therapeutically effective amount of one or more compound of the invention together with one or more anti-cancer therapeutics. The compound(s) can be administered before, during or after treatment with the anti-cancer therapeutic. An "anti-cancer therapeutic" is a compound, composition or treatment that prevents or delays the growth and/or metastasis of cancer cells. Such anti-cancer therapeutics include, but are not limited to, chemotherapeutic drug treatment, radiation, gene therapy, hormonal manipulation, immunotherapy and antisense oligonucleotide therapy. A wide variety of chemotherapeutic drugs are known in the art and can be used in combination therapies with a compound of the present invention. Examples of useful chemotherapeutic drugs include broad spectrum chemotherapeutics, i. e. those that are useful in the treatment of a range of cancers, such as doxorubicin, capecitabine, mitoxantrone, irinotecan (CPT-11), cisplatin and gemcitabine. Other examples of useful chemotherapeutic agents include, but are not limited to, hydroxyurea, busulphan, carboplatin, chlorambucil, melphalan, cyclophosphamide, Ifosphamide, danorubicin, epirubicin, vincristine, vinblastine, Navelbine® (vinorelbine), etoposide, teniposide, paclitaxel, docetaxel, cytosine, arabinoside, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C and the like. The compounds of the invention are also suitable for use with standard combination therapies employing two or more chemotherapeutic agents. It is to be understood that anti-cancer therapeutics for use in the present invention also include novel compounds or treatments developed in the future.
  • VI. Pharmaceutical Compositions
  • The compounds of the present invention are typically formulated prior to administration. The present invention thus provides pharmaceutical compositions comprising one or more compounds of the invention and a pharmaceutically acceptable carrier, diluent, or excipient. The pharmaceutical compositions are prepared by known procedures using well-known and readily available ingredients. Pharmaceutical compositions comprising one or more compounds of the invention in combination with one or more known cancer chemotherapeutics are also contemplated by the present invention.
  • Compounds of the invention or pharmaceutical compositions comprising the compounds may be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. In the usual course of therapy, the active compound is incorporated into an acceptable vehicle to form a composition for topical administration to the affected area, such as hydropohobic or hydrophilic creams or lotions, or into a form suitable for oral, rectal or parenteral administration, such as syrups, elixirs, tablets, troches, lozenges, hard or soft capsules, pills, suppositiories, oily or aqueous suspensions, dispersible powders or granules, emulsions, injectables, or solutions. The term parenteral as used herein includes subcutaneous injections, intradermal, intra-articular, intravenous, intramuscular, intravascular, intrasternal, intrathecal injection or infusion techniques.
  • Compositions intended for oral use may be prepared in either solid or fluid unit dosage forms. Fluid unit dosage form can be prepared according to procedures known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. An elixir is prepared by using a hydroalcoholic (e.g., ethanol) vehicle with suitable sweeteners such as sugar and saccharin, together with an aromatic flavoring agent. Suspensions can be prepared with an aqueous vehicle with the aid of a suspending agent such as acacia, tragacanth, methylcellulose and the like.
  • Solid formulations such as tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate: granulating and disintegrating agents for example, corn starch, or alginic acid: binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc and other conventional ingredients such as dicalcium phosphate, magnesium aluminum silicate, calcium sulfate, starch, lactose, methylcellulose, and functionally similar materials. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate maybe employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. Soft gelatin capsules are prepared by machine encapsulation of a slurry of the compound with an acceptable vegetable oil, light liquid petrolatum or other inert oil.
  • Aqueous suspensions contain active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxylmethylcellulose, methyl cellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia: dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example hepta-decaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl- p-hydroxy benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example peanut oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavouring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present.
  • Pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oil phase may be a vegetable oil, for example olive oil or peanut oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
  • The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or a suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Adjuvants such as local anaesthetics, preservatives and buffering agents can also be included in the injectable solution or suspension.
  • The compound(s) of the invention may be administered, together or separately, in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.
  • Other pharmaceutical compositions and methods of preparing pharmaceutical compositions are known in the art and are described, for example, in "Remington: The Science and Practice of Pharmacy" (formerly " Remingtons Pharmaceutical Sciences"); Gennaro, A., Lippincott, Williams & Wilkins, Philidelphia, PA (2000).
  • VII. Administration of Compounds of the invention
  • Compounds of the invention may be administered to a subject by a variety of routes depending on the cancer to be treated, for example, the compounds may be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations. In one embodiment, the compounds are administered systemically to a subject, for example, by bolus injection or continuous infusion into a subject's bloodstream or by oral administration. When used in conjunction with one or more known chemotherapeutic agents, the compounds can be administered prior to, or after, administration of the chemotherapeutic agents, or they can be administered concomitantly. The one or more chemotherapeutic may also be administered systemically, for example, by bolus injection, continuous infusion, or oral administration.
  • The compounds of the invention may be used as part of a neo-adjuvant therapy (to primary therapy), or as part of an adjuvant therapy regimen, where the intention is to cure the cancer in a subject. The present invention contemplates the use of the compounds of at various stages in tumour development and progression, including in the treatment of advanced and/or aggressive neoplasias (i.e. overt disease in a subject that is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy), metastatic disease, locally advanced disease and/or refractory tumours (i.e. a cancer or tumour that has not responded to treatment).
  • "Primary therapy" refers to a first line of treatment upon the initial diagnosis of cancer in a subject. Exemplary primary therapies may involve surgery, a wide range of chemotherapies and radiotherapy. "Adjuvant therapy" refers to a therapy that follows a primary therapy and that is administered to subjects at risk of relapsing. Adjuvant systemic therapy is usually begun soon after primary therapy to delay recurrence, prolong survival or cure a subject.
  • It is contemplated that the compounds of the invention can be used alone or in combination with one or more other chemotherapeutic agents as part of a primary therapy or an adjuvant therapy. Combinations of the compounds of the invention and standard chemotherapeutics may act to improve the efficacy of the chemotherapeutic and, therefore, can be used to improve standard cancer therapies. This application can be important in the treatment of drug-resistant cancers which are not responsive to standard treatment. Drug-resistant cancers can arise, for example, from heterogeneity of tumour cell populations, alterations in response to chemotherapy and increased malignant potential. Such changes are often more pronounced at advanced stages of disease.
  • The dosage to be administered is not subject to defined limits, but it will usually be an effective amount. It will usually be the equivalent, on a molar basis of the pharmacologically active free form produced from a dosage formulation upon the metabolic release of the active free drug to achieve its desired pharmacological and physiological effects. The compositions may be formulated in a unit dosage form. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Examples of ranges for the compound(s) in each dosage unit are from about 0.05 to about 100 mg, or more usually, from about 1.0 to about 50 mg.
  • Daily dosages of the compounds of the present invention will typically fall within the range of about 0.01 to about 100 mg/kg of body weight, in single or divided dose. However, it will be understood that the actual amount of the compound(s) to be administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms. The above dosage range is given by way of example only and is not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing harmful side effects, for example, by first dividing the larger dose into several smaller doses for administration throughout the day.
  • VIII. Kits
  • The described herein are therapeutic kits containing one or more compounds of Formula VI for use in the treatment of cancer. The contents of the kit can be lyophilized and the kit can additionally contain a suitable solvent for reconstitution of the lyophilized components. Individual components of the kit would be packaged in separate containers and, associated with such containers, can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, for use or sale for human or animal administration. When the components of the kit are provided in one or more liquid solutions, the liquid solution can be an aqueous solution, for example a sterile aqueous solution. For in vivo use, the compounds may be formulated into a pharmaceutically acceptable syringeable composition. In this case the container means may itself be an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the formulation may be applied to an infected area of the subject, such as the lungs, injected into an subject, or even applied to and mixed with the other components of the kit. Pharmaceutical kits or packs comprising one or more compound of the present invention in combination with one or more standard chemotherapeutic for combination therapy applications are also contemplated by the present invention.
  • To gain a better understanding of the invention described herein, the following examples are set forth. It should be understood that these examples are for illustrative purposes only. Therefore, they should not limit the scope of this invention as defined by the appended claims in any way.
  • EXAMPLES Preparation of compounds of the invention and of reference compounds:
  • All reactions have been carried out according to the scheme shown below:
    Figure imgb0056
    • R2 and R3 are independently aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl or R2 and R3 when taken together along with the carbon atoms they are attached to, form aryl or substituted aryl, heterocycle, substituted heterocycle, heteroaryl, or substituted heteroaryl; and
    • R4 is hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkyl alkynyl, alkoxy, alkylthio, substituted alkylthiol, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, cyano, -S(O)0-2R wherein R is alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl.
  • In a typical experimental procedure 1 mmol (1 equiv.) of the carboxyaldehyde was combined with 1.05 - 1.10 mmole (1.05 - 1.1 equiv.) of the dione and 20 mmole (20 equiv.) of ammonium acetate and 5 ml of acetic acid. The mixture was magnetically stirred and heated to reflux for 3-5 hr. The reaction process was monitored by TLC,ntil complete consumption of the indole was achieved. The reaction mixture was cooled to room temperature and added drop-wise into well-stirred ice-water. The suspension solid was then filtered and the crude solid was dissolved in ethyl acetate, dried over sodium sulfate and filtered, the organic solvent was removed by vacuum. The products was then either recrystalized with alcohol or separated by column chromatography using petroleum ether-Ethyl acetate as an eluant.
  • Melting points were recorded using a MEL-TEMP capillary melting point apparatus, the melting point are uncorrected. 1H-NMR was performed in a 500 MHz Brucker instrument at room temperature using a suitable deuterated solvent.
  • The following exemplary compounds were prepared from the appropriate starting materials following the general synthetic procedure as discussed above.
  • Example 28: Compound 88
  • Figure imgb0057
  • Mp 335-336 °C. 1H-NMR (DMSO-d6), two isomers: 1) δ =13.160 (s) 1H, 11.602 (s) 1H, 9.720 (s) 1H, 9.143 (dd) 1H, 8.975 (dd) 1H, 8.680 (d) 1H, 8.345 (t) 1H, 8.160 (d) 1H, 7.870 (t) 1H, 7.720 (t) 1H, 7.420 (d) 1H, 7.200 (d) 2H, 2.862 (s) 3H: δ =13.090 (s) 1H, 11.602 (s) 1H, 9.370 (s) 1H, 9.143 (dd) 1H, 8.975 (dd) 1H, 8.680 (d) 1H, 8.345 (t) 1H, 8.099 (d) 1H, 7.870 (t) 1H, 7.720 (t) 1H, 7.420 (d) 1H, 7.200 (d) 2H, 2.847 (s) 3H. EI-MS (C24H16N4O2) = 392.
  • Example 29: Compound 90
  • Figure imgb0058
  • 1H-NMR (DMSO-d6): δ =13.083 (s) 1H, 11.595 (s) 1H, 9.040-9.010 (m) 4H, 8.950 (d) 1H, 8.120 (m) 1H, 7.821 (t) 1H, 7.432 (m) 1H, 7.176 (m) 2H, 2.830 (s) 3H.
  • Example 31: Compound 94
  • Figure imgb0059
  • Mp 300-303 °C. 1H-NMR (DMSO-d6): δ = 12.55 (s) 1H, 8.83 (m) 3H, 8.68 (m) 1H, 8.50 (m) 1H, 8.15 (m) 1H, 7.75 (m) 2H, 7.65 (m) 1H, 7.47 (m) 1H, 7.40 (m) 1H. ESI-MS (C23H15BrN4) = 427.
  • Example 33: Compound 97
  • Figure imgb0060
  • 1H-NMR (DMSO-d6): δ =13.460 (s) 1H, 11.890 (s) 1H, 9.080-8.985 (m) 4H, 8.860-8.825 (m) 1H, 8.285 (d) 1H, 7.890-7.840 (m) 2H, 7.560-7.540 (m) 1H, 7.420-7.390 (m) 1H. ESI-MS (C21H12BrN5) = 414.
  • Example 34: Compound 101
  • Figure imgb0061
  • EI-MS : 484.00 (C25H18BrN5O requires 483.07) H1 NMR (DMSO-d6) d= 13.167 (s) 1H, 11.845 (s) 1H, 8.839-8.781 (m) 4H, 8.292-8.405 (m) 2H, 7.364-7.697 (m) 5H, 3.543 (s) 1H, 3.410 (s) 2H, 1.463 (s) 2H.
  • Example 37: Compound 146
  • Figure imgb0062
  • Mp 240-242 °C. 1H-NMR (DMSO-d6): δ = 12.20 (s) 1H, 11.90 (s) 1H, 8.85 (m) 2H, 8.60 (d) 1H, 8.40 (d) 1H, 7.70 (m) 8H, 7.30 (m) 5H.
  • Example 39: Compound 156
  • Figure imgb0063
  • Mp 365-366 °C 1H-NMR (DMSO-d6), two isomers: a) δ = 13. 410 (s) 1H, 11.670 (s) 1H, 9.40 (d) 1H, 9.11 (d) 1H, 8.96 (d) 1H, 8.70 (d) 1H, 8.35-8.18 (m) 2H, 7.96 (s) 1H, 7.70 (t) 1H, 7.56 (t) 1H, 7.28 (m) 2H.
    b) δ = 13.290 (s) 1H, 11.67 (s) 1H, 9.305(d) 1H, 9.095 (d) 1H, 8.960 (d) 1H, (d) 1H, 8.70 (d) 1H, 8.495 (d) 1H, 7.87 (d) 1H, 7.70 (t) 1H, 7.56 (t) 1H, 7.28 (m) 2H. EI-MS (C23H14N4O2) = 378.
  • Example 40: Compound 157
  • Figure imgb0064
  • 1H-NMR (DMSO-d6): δ =13.102 (s) 1H, 11.702 (s) 1H, 8.900-8.840 (m) 3H, 8.690 (d) 1H, 8.445-8.400 (m) 3H, 8.301 (d) 1H, 7.747 (t) 1H, 7.644-7.624 (m) 1H, 7.622-7.605 (m) 1H, 7.585-7.529 (m) 1H, 7.128-7.086 (m) 1H. EI-MS (C23H14N3F) = 351.
  • Example 43: Compound 169
  • Figure imgb0065
  • Mp 236-237 °C. EI-MS: 633.87 (C32H19Br2N5) require 633.33. 1H-NMR (DMSO-d6), two isomers: 1) δ =13.190 (s) 1H, 11820 (s) 1H, 8.955 (s) 1H, 8.910-8.883 (m) 3H, 8.640 (d) 1H, 8.590 (d) 1H, 8.280 (d) 2H, 8.157 (d) 1H 7.730
    (t) 1H, 8.620 (t) 1H, 7.525 (s) 1H, 7.509 (s) 1H, 7.410 (d) 1H, 7.375 (d) 1H.
    2) δ =13.190 (s) 1H, 12.060 (s) 1H, 8.955 (s) 1H, 8.910-8.883 (m) 3H, 8.640 (d) 1H, 8.590 (d) 1H, 8.280 (d) 2H, 8.157 (d) 1H 7.730
    (t) 1H, 8.620 (t) 1H, 7.525 (s) 1H, 7.509 (s) 1H, 7.410 (d) 1H, 7.375 (d) 1H.
  • Example 44: Compound 175
  • Figure imgb0066
  • EI-MS: 440.11 (C25H18BrN3 requires 440.07. 1H-NMR (DMSO-d6): δ =13.141 (s) 1H, 11.779 (s) 1H, 8.842 (d) 1H, 8.673 (s) 1H, 8.636 (s) 1H, 8.508 (d) 1H, 8.295 (d) 1H, 8.242 (d) 1H, 7.553 (d) 2H, 7.508 (d) 1H, 7.365 (d) 1H, 2.612 (s) 6H.
  • Example 49: In vitro Inhibition of Proliferation of Cancer Cells #1
  • Selected compounds of the invention were tested for anti-cancer activity in vitro using a human colon carcinoma cells (HT-29) and human non-small cell lung cancer cells (H460). The cells were maintained in α-MEM medium (Wisent, St-Bruno, Qc) supplemented with 10% FBS, and grown at 37°C in an atmosphere of 5% CO2. Cells were transferred onto 150mm tissue culture plates and grown until sub-confluency (70-80%) prior to their use.
  • The anti-cancer activity in vitro was evaluated by a cell proliferation assay based on the ability of live cells to reduce the tetrazolium salt XTT to orange coloured compounds of formazan (XTT cell proliferation kit II, Roche Applied Science, Montreal, QC).
  • Approximately 4 x 103 colon cancer cells (HT-29) or 2 x 103 non-small cell lung cancer cells (NCI-H460) in 100 µl of complete culture medium were plated onto 96-well microtiter plates and incubated overnight at 37°C. The medium was then removed by inverting the plate and patting on a sterile absorbent cloth. Fifty µl of medium containing the test compound at either 25 or 100 µM, were added to the wells containing cells and incubated at 37°C in an atmosphere of 5% CO2 for 48 h. Following incubation, 25 µl of an XTT reaction mixture (XTT at a final concentration of 0.3 mg/ml) were added to each well and the plates were incubated for a further 4 h. The absorbance of each sample was then determined at a wavelength of 490 nm/650 nm as reference. Each compound was tested in duplicate and the results are reported as averages. Table II shows the effect that different compounds of Formula I have on the growth of human colon carcinoma HT-29. Table III shows the effect that different compounds of Formula I have on the growth of human non-small cell lung cancer cells (H460). Table II: Inhibition of Proliferation of Human Colon Carcinoma (HT-29) Cells
    Compound 100µM 25µM
    % Survival SD(%) % Survival SD(%)
    44 6.5 0.4 63.6 2.9
    45 35 0.6 88.9 3.3
    46 4.5 0 16.1 1
    CPT-11 51.1 3.2 82.3 10
    Vehicle 100 7 100 7
    Table III: Inhibition of Proliferation of Human Lung Carcinoma (NCI- 460) Cells
    Compound 100µM 25µM
    % Survival SD(%) % Survival SD(%)
    44 3.4 1 77.3 5.3
    45 32.7 5.5 96 1.1
    46 1.9 0.1 11.5 1.4
    CPT-11 6.1 0.5 32.2 4.5
    Vehicle 100 3.4 100 3.4
  • Example 50: In vitro Inhibition of Proliferation of Cancer Cells #2
  • The compounds listed below were tested for anti-cancer activity against several carcinoma cell lines as described below and in Examples 51-53. Examples 94, 97, 99, 100, 101, 102, 103, 104, 105 and 106 are in accordance with the present invention.
    Figure imgb0067
    Figure imgb0068
    Figure imgb0069
    Figure imgb0070
    Figure imgb0071
  • Cells were maintained in α-MEM medium (Wisent, St-Bruno, QC) supplemented with 10% FBS, and grown at 37°C in an atmosphere of 5% CO2. They were transferred onto 150mm tissue culture plates and grown until sub-confluency (70-80%) prior to their use.
  • The anti-cancer activity in vitro was evaluated by a cell proliferation assay based in the ability of live cells to reduce the tetrazolium salt XTT to orange coloured compounds of formazan (XTT cell proliferation kit II, Roche Applied Science, Montreal, QC). The following cancer cell lines were tested: HT-29 colon carcinoma, A498 renal carcinoma, Caki-1 renal carcinoma, C8161 melanoma, MDA-MB-231 breast adenocarcinoma, A2058 metastatic melanoma, SK-OV-3 ovarian adenocarcinoma, Hep G2 liver carcinoma, AsPC-1 pancreatic adenocarcinoma, PC3 metastatic prostate adenocarcinoma. WI 38 is a human lung fibroblast cell line.
  • Approximately 2-3×103 cells in 100 µl of complete culture medium were plated onto 96-well microtiter plates and incubated overnight at 37°C, the medium was removed by inverting plate and patting on sterile absorbent cloth. Fifty µl of medium containing the different compounds at different concentrations were added and wells were incubated at 37°C with 5% CO2 for 48 h. Following incubation, 25 µl of an XTT reaction mixture (XTT at a final concentration of 0.3 mg/ml) were added and wells were incubated for 4 h. The absorbance of each sample was determined at a wavelength of 490nm/650 nm as reference. The percentage of survival was determined by the ratio between absorbance values of cells incubated with the different compounds and their respective controls (cells incubated with vehicle only). The results are shown in Figures 1-4.
  • Figure 1 depicts the results with compound 92; Figure 2 depicts the results with compound 28; Figure 3 depicts the results with compound 50; and Figure 4 depicts the results with compound 42.
  • Example 51: Concentration Dependence of Inhibition of Cancer Cell Proliferation by Compound 45 in vitro
  • The effect of various concentrations of compound 45 on various cancer cell lines was tested following the general protocol outlined in Example 50, with the following exceptions. Cell survival was assessed 48 h, 72 h and 6 days post-treatment by incubating cells with XTT for 2 h. The cancer cell lines utilised in this example were the same as those listed in Example 50, together with the cervical carcinoma cell line KB. The results are shown in Figures 5 and 6, which depict cell survival after treatment with various concentrations of compound 45. A. 48 h after treatment, B. 72 h after treatment and C. 6 days after treatment.
  • Example 52: Concentration Dependence of Inhibition of Cancer Cell Proliferation by Compounds 45 and 99 In Vitro
  • The effect of various concentrations of compounds 45 and 99 on the colon carcinoma cancer cell line LS513 was tested following the general protocol outlined in Example 50, with cell survival being assessed 6 days post-treatment. The results are shown in Figure 7.
  • Example 53: In Vitro Inhibition of Proliferation of Colon Carcinoma Cells #1
  • The effect of various compounds of the invention on the proliferation of HT-29 colon carconoma cells was tested following the general protocol outlined in Example 50 with the exception that cell survival was assessed after 5 to 7 days of treatment. The results using concentrations of 2, 10 and 25µM of each compound are shown in Figure 8 ( Compounds 94 and 100 to 106 are compounds of the invention). Results were compiled from different experiments with 5 to 7 days of treatment. The coefficient of variation for most samples were within 5%.
  • Example 54: In Vitro Inhibition of Proliferation of Cancer Cells #3
  • The twenty-three compounds shown below were evaluated for their antiproliferative effects in a panel of 60 human cancer cell lines as part of the in vitro anticancer screening services provided by the DTP (Developmental Therapeutics Program) of the US National Cancer Institute (NCI) U.S (Compounds 88, 89, 90, 45, 94 and 97 are compounds of the invention). National Cancer Institute (NCI) of the National Institutes of Health (NIH) in Rockwell, Maryland. The cancer cell lines used in this screen are provided in Figure 9.
    Figure imgb0072
    Figure imgb0073
    Figure imgb0074
    Figure imgb0075
    Figure imgb0076
  • The NCI conducts a standard 48/72 hour 60 cell line assay and an in vitro time course assay as described in Alley et al. (Cancer Res (1988) 48:589). In the standard 60 cell line assay, a minimum of 5 concentrations of the test compound are tested at 10-fold dilutions against 60 cell lines and cell growth is assayed at 48 and 72 hours using a sulphorhodamine B assay. For the time course analysis, tumour cells are treated with the test compound at various time points, then washed and grown in medium free of the test compound until the end of the experiment at 144 hrs. This assay employs 20% FBS to better approximate the minimum c x t (concentrations and times) test compound exposure conditions that are required to achieve activity in vivo. Cell growth is quantified by an MTT assay (similar to the XTT assay described above) and the concentration of the test compound required for growth inhibition is determined. The inhibitory effect of the test compounds are expressed as a GI50 value, which represents the molar concentration of the test compound that results in 50% growth inhibition.
  • All compounds exhibited antiproliferative activity against all human tumour cell lines including NSCLC, leukemia, colon cancer, prostate cancer, melanoma, ovarian cancer, renal cancer, CNS cancer, and breast cancer, with GI50 (growth inhibition by 50%) values ranging from 0.61 µM to 12.3 µM, with an average of 2 µM. The compound 45 had a GI50 value of 2.0 µM, while the most effective compound was 90 (Figure 10A). The compounds affected the growth of all cell lines comparatively equally. The average GI50 values for compound 45 ranged from 1.3 µM (renal) to 3.4 µM (leukemia) (Figure 10B). These results suggest that compound 45 affects a ubiquitous target. The TGI (total growth inhibition) for this compound towards leukemia cell lines was significantly different from that of other cell types. These cell lines were not 100% growth inhibited, even at 100 µM, the highest concentration used (Figure 10C).
  • Example 55: In Vitro Inhibition of Proliferation of Lung Cancer Cells
  • The following compounds were tested for theier ability to inhibit the proliferation of H460 non-small cell lung carcinoma cells in vitro. The protocol described in Example 50 was utilised with the exception that cell survival was assessed after 6 days of treatment. Each compound was tested at concentrations of 0.2, 2, 10 and 25 µM. The results are shown in Figure 11 ( Compounds 101, 103 and 113 are compounds of the invention).
    Figure imgb0077
    Figure imgb0078
    Figure imgb0079
  • Example 56: In Vitro Inhibition of Proliferation of Colon Carcinoma Cells #2
  • The above compounds (as shown in Example 55), together with those shown below, were tested for their ability to inhibit the proliferation of HT-29 colon carcinoma cells in vitro. The protocol described in Example 50 was utilised with the exception that cell survival was assessed after either 2 or 6 days of treatment. Each compound was tested at concentrations of 0.2, 2, 10 and 25 µM ( compounds 110, 30, 101,113, 103, 107, 108 and 109) or at concentrations of 2.5, 10 and 25 µM ( compounds 112, 114, 78, 111 and 45). The results are shown in Figures. 12 and 13. The results shown in Figure 12 reflect cell survival 6 days after treatment with the listed compounds. Figure 13A shows cell survival 2 days after treatment with the listed compounds and Figure 13B shows cell survival 6 days after treatment. Compounds 101, 113, 45 and 114 are compounds of the invention.
    Figure imgb0080
    Figure imgb0081
  • Example 57: Inhibition of Colon Carcinoma Growth In Vivo #1
  • This Example and the following Example 58 describe in vivo efficacy studies of various compounds of the invention performed using a mouse xenograft model using the human colon adenocarcinoma cell line HT-29. The following compounds were tested ( Compounds 44, 45 and 46 are compounds of the invention).
    Figure imgb0082
    Figure imgb0083
    Figure imgb0084
    Figure imgb0085
  • Groups of five to 10 CD-1 female nude mice (6-7 weeks) were injected in the lower mid back with human colon adenocarcinoma cells HT-29 (3 x 106 cells in 0.1 ml PBS) subcutaneously, and the treatment initiated 5 days post-inoculation (size of tumours = 20-40 mm3). The treatment schedule consisted of 2 x 200 µl intraperitoneal injections per day of 5 mg/ml (100 mg/Kg/d) for five days and 2 days break, for 4 weeks. Tumour sizes were measured during the course of the treatment using calipers, mice were then sacrificed by cervical dislocation and tumours surgically removed and weighed. Figures 14 shows the average tumour size (mm3) in the different groups of mice. Figures 15 and 16 show the average tumour weight per group of mice and per individual mouse, respectively.
  • Example 58: Inhibition of Colon Carcinoma Growth In Vivo #2
  • The protocol described in Example 55 was followed. The results are shown in Figure 17, which depicts the average tumour size (mm3) in the different groups of mice. Abbreviations used in Figure 17 are as follows: V-ip = Vehicle (i.p); 42 (5-ip) = 5 mg/Kg (i.p.); 42 (25-1p)= 25 mg/Kg; 42 (100-ip); 100 mg/Kg (i.p.); 43 (ip) = 100 mg/Kg(i.p.); 45 (ip)= 100 mg/Kg (i.p); 44(ip)= 100 mg/Kg(i.p.); 46 = 100 mg/Kg (i.p); 28= 100 mg/kg (i.p); V-op = vehicle, oral; 42 (100-op) = 100 mg/Kg (oral).
  • Example 59: In Vivo Inhibition of Cancer Cell Growth by Compound 45
  • The ability of compound 45 to inhibit the growth of cancer cells in vivo was further investigated in a mouse xenograft model of hepatocellular (liver) cancer. Groups of five to 10 CD-1 female nude mice were injected subcutaneously in the mid right flank with HepG2 human hepatocarcinoma cells (1 x 107 cells). The treatment was initiated 7 days post-inoculation and consisted of 2 x 200 µl intraperitoneal injections per day (100 mg/Kg/d). Tumour sizes were measured during the course of the treatment using calipers, and were surgically removed and weighed after 10 weeks. The results obtained are shown in Figure 18A & B.
  • Notably, none of the compounds tested in the preceding Examples 57-59 showed toxic effects in vivo.
  • Example 60: Effect of Compound 45 on the Activity of Various Human Kinase Enzymes #1
  • Compound 45 was tested for its ability to function as a kinase inhibitor using the kinase profiler service from Upstate Biotechnologies. The general protocol employed is as follows: recombinant kinases were incubated with specific substrates, 10 mM MgAcetate, and [γ-33P-ATP]. The reaction was initiated by the addition of MgATP mix. After incubation at room temperature for 40 minutes, the reaction was stopped by the addition of 5 µl of a 3% phosphoric acid solution. 10 µl of the reaction was then spotted on to a P30 filtermat and washed 3 times for 5 min. in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. Each reaction was performed in duplicate with 100 µM ATP -/+ 10 µM compound 45. Results are presented in Table IV and are expressed as the mean of % control (no compound). PI 3-kinase-y (PI3K-γ) activity was determined with the PIProfiler™ assay, which measures the binding of the GRP1 pleckstrin homology (PH) domain to PIP3, the product of PI3K acting on its physiological substrate PIP2.
  • Seventy-nine recombinant kinases were tested. Of these 87% retained greater then 60% activity in the presence of 10 µM ML-220. Four kinases retained between 40 and 60% activity (Alk-60%; Aurora-A, 54%; PKD2, 52%; SAPK3, 54%; TrkA, 56%), whereas 5 kinases had less than 40% activity (CaMKII, 32%; PI3Kα, 30%; PI3Kβ, 11%; PI3Kδ, 9%; and PI3Kγ, 22%). These results indicate that compound 45 can function as a kinase inhibitor, and it has a high degree of selectivity for particular kinases. Table IV: Kinase Inhibiting Activity of Compound 45
    Kinase Family % activity
    Abl TK
    107
    ALK TK 60
    AMPK CAMK
    100
    ASK1 STE 99
    Aurora-A other 54
    Axl TK 100
    BRK TK 112
    CaMKII CAMK 32
    CaMKIV CAMK 98
    CDK1/cyclinB CMGC 156
    CDK2/cyclinA CMGC 95
    CDK2/cyclinE CMGC 117
    CDK3/cyclinE CMGC 107
    CDK6/cyclinD3 CMGC 87
    CDK7/cyclinH/MAT1 CMGC 95
    CHK1 CAMK 111
    CK2 other 91
    EGFR TK 105
    EphA2 TK 95
    EphB4 TK 95
    ErbB4 TK 73
    Fes TK 99
    FGFR3 TK 82
    Fms TK 135
    Fyn TK
    103
    GSK3α CMGC
    96
    IGF-1R TK 80
    IKKβ other 111
    IKKα other 150
    JNK1α1 CMGC 85
    JNK3 CMGC 121
    Lyn TK 81
    MAPK1 CMGC 85
    MAPK2 CMGC 99
    MAPKAP-K2 CAMK 119
    MEK1 STE 88
    Met TK 129
    MINK STE 91
    MKK4 STE 96
    MKK6 STE 86
    MSK1 AGC 76
    MST2 STE 77
    NEK2 other 90
    p70S6K AGC 64
    PAK2 STE 89
    PAR-1Bα CAMK 88
    PDGFRα TK 117
    PDK1 AGC
    106
    PI3K□ LIPID 22
    PI3K-β LIPID 11
    PI3K-α LIPID 30
    PI3K-δ LIPID 9
    Pim-1 CAMK 70
    PKA AGC 83
    PKBα AGC 95
    PKCµ AGC 90
    PKCα AGC 92
    PKCδ AGC 87
    PKCζ AGC 96
    PKD2 CAMK 52
    Plk3 other 132
    PRK2 AGC 83
    RAF TKL 100
    Ret TK 82
    ROCK-II AGC 86
    Ros TK 107
    Rse TK 176
    Rsk1 AGC 183
    SAPK2a CMGC 62
    SAPK2b CMGC 80
    SAPK3 CMGC 54
    SAPK4 CMGC 77
    SGK AGC 89
    SRC TK 102
    TAK1 TKL 104
    Tie2 TK 109
    TrkA TK 56
    Yes TK 91
  • Example 61: Effect of Other Compounds on the Activity of Various Human Kinase Enzymes
  • To assess whether other compounds also affected the same kinases, the inhibitory activity of 10 µM of compound 90 was tested on five kinases: Aurora-A, CaMKII, PKD2, SAPK3, TrkA and PI3K. The results are shown in Figure 19. The results indicated that the compound has a different pattern of kinase inhibition than compound 45.
  • Example 62: Determination of the Subcellular Localization of Compound 45 in Various Cancer Cells
  • Compound 45 is intrinsically fluorescent, which allowed the subcellular localization of this compound to be examined by fluorescent microscopy. Fluorescent microscopy was performed at the Microscopy Imaging Centre, Faculty of Medicine, University of Toronto. Cells were treated with 100 µM of compound 45 (Figure 20A, B, D, E) or 1 µM doxorubicin (Figure 20C) for 1 hour, washed once in PBS, fixed in 3.7% formaldehyde/PBS for 10 minutes, washed three times in PBS and mounted with Immuno-fluoro. Images were obtained with a Zeiss laser scanning fluorescent microscope with an excitation filter range of 360-370 nm (compound 45) or 530-560 nm (doxorubicin). For Figure 20B and C, differential interference contrast images were overlaid with fluorescent images.
  • Compound 45 localizes to punctuate spots in the perinuclear area of HT-29 colon adenocarcinoma cells (Figure 20A), and is excluded from the nucleus and plasma membrane regions (Figure 20B). In contrast, the anti-cancer agent, doxorubicin, which is also intrinsically fluorescent, is localized in the nucleus (Figure 20C). A similar localization for compound 45 was observed in A498 renal carcinoma cells (Figure 20D) and C8161 melanoma cells (Figure 20E).
  • Example 63: Determination of Morphological Changes in Cells Treated with Compound 45
  • Treatment with compound 45 for 24 hours leads to the formation of large vacuoles within the cytoplasm of HT29 colon adenocarcinoma cells (Figure 21), A498 renal carcinoma cells and MDA-MB-231 breast adenocarcinoma cells. These vacuoles are not formed in DMSO- or doxorubicin-treated cells. Moreover, the nuclear membrane is no longer evident in the phase-contrast images of cells treated with compound 45, even though the nucleus is still intact, as shown by DAPI staining. Figure 21 shows differential interference contrast (DIC) images (top row) and fluorescent images (lower row) of the same cells stained with DAPI, a. cell permeable marker for the nucleus.
  • Example 64: Cell Cycle Analysis
  • The effect of treatment with compound 45 on cell cycle progression in HT-29 colon adenocarcinoma cells was examined by flow cytometry (Figure 22). Values were determined by gate analysis of flow cytometric plots and are presented in Figure 22 as a percentage of the total cell population, after eliminating doubles. Apoptotic events inferred by the surface area preceding G1 phase. Cells were starved for 3 days, and treated with 15 µM or 25 µM compound 45 for 24 or 48 hours in the presence of 10% serum, followed by flow cytometric analysis. Treatment with compound 45 led to an increase of cells in the G1 phase and a decrease in the S and G2/M phases.
  • The results presented above in this Example and in Examples 58, 60 and 61 indicate that compound 45 suppressed the growth of HT-29 colon cancer cells with a GI50 of 2.6 µM, and induced a partial arrest in the G0/G1 phase of the cell cycle. Fluorescent microscopy revealed the presence of compound 45 within the cytoplasm, but not the nucleus or plasma membrane regions of the cell. In addition, compound 45 was found to inhibit kinase activity in a screen of protein kinases, indicating that the cellular target may be a cytoplasmic protein kinase. These results indicate that compound 45 and related derivatives have potential as therapeutic agents for the treatment of human cancer.
  • Example 65: Selectivity of Compounds of the invention
  • Compounds of the invention that demonstrate the ability to decrease the growth or proliferation of at least one cancer cell line may undergo further testing to evaluate their selectivity towards cancer cells. An exemplary method to measure the selectivity of the compounds of the present invention is provided below.
  • IC90 values of selected compounds on a panel of normal actively proliferating cells (HUVEC and WI38) and cancer cells representing colon (HT-29), lung (NCI-H460), breast (MDA-MB-231) and prostate cancer (PC-3) are measured. Compounds with 2-fold or higher overall selectivity to the panel of cancer cell lines at IC90 are identified as potential therapeutics.
  • IC90 values are determined using the XTT assay as an indicator of growth arrest and/or cytotoxicity. This assay is conducted as outlined in Example 50. Percentage inhibition is calculated for each cell line and IC90 values for each compound and cell type determined. The average IC90 values for the normal cells are calculated and divided by the average IC90 values for the cancer cell lines. Compounds with a selectivity ratio of >2 are identified and chosen for further optimization and/or testing.
  • Example 66: Additional In Vivo Anti-tumour Efficacy Evaluations
  • Further pharmacological evaluation of selected compounds is conducted in animal models of human tumour growth. Data from these studies provide evidence of the therapeutic efficacy of selected compounds against various types of cancer and help to identify compounds with better pharmacological properties and potency.
  • Examples of mouse models that can be utilized to investigate the efficacy of selected compounds include, but are not limited to, xenografts of various human tumour types, inoculated subcutaneously into nude mice or mice with severe combined immunodeficiency disorder (SCID) as described above; orthotopic implantation of various human tumours in nude or SCID mice for investigation of effects on the tumour in the target organ (for example, a pancreatic cancer cell graft implanted directly into the pancreas of the animal), and investigation of spontaneous tumours in normal mice.
  • In order to provide evidence of the efficacy of a selected compound as a single agent, it may be evaluated, for example, in specific models (xenograft or orthotopic) for representative human cancers such as pancreas, skin (melanoma), kidney, colon, breast, lung, liver, ovary, prostate, bladder and brain. Similar studies can be conducted to evaluate the performance of test compounds in combination with other standard therapeutic modalities used in the treatment of human cancers.
  • For typical xenograft studies, 5-6 week old, female, CD-1 athymic nude mice, (Charles River, Montreal, QC) are acclimatized in a pathogen-free facility for at least 1 week. Animal protocols followed are in compliance with the Guide for the Care and Use of Laboratory Animals in Canada. Approximately 106-107 human tumour cells in 100 ml PBS are subcutaneously injected into the right flank of each mouse. Once tumours reach an approximate volume of 100 mm3 (several days post tumour cell injection), mice are randomized by tumour size into control and treatment groups. Test compounds are administered at various doses 5 days a week for several weeks. Control animals receive vehicle alone (negative control) and/or a standard chemotherapeutic (positive control) for the same period. The tumour dimensions (length, width, and height) are measured using calipers twice a week over the treatment period. Tumour volume is calculated by the formula L x W x H/2, where L indicates length, W indicates width and H indicates height. The mice are sacrificed when the tumour burden reaches approximately 10% of total body weight and excised tumours are weighed. A standard bar graph is used to demonstrate the differences in tumour weights with each bar representing mean tumour weight.
  • Example 67: Additional Assays to Investigate Potential Mechanism of Action
  • The potential mechanism of action of selected compounds can be investigated using assays such as cell-cycle analysis, apoptosis assays, anti-angiogenesis assays and immunohistochemical analysis. A representative example of each type of assay is provided below.
  • i) Cell-cycle analysis
  • Alterations in cell cycle are determined using flow cytometric analyses. Tumour cells sensitive to a test compound are synchronized by plating in medium containing 0.5% FBS for 24 h followed by culturing in FBS-free medium for 48 h. The cells are then released into complete medium containing 0.1% DMSO (vehicle control) or the test compound at an appropriate concentration (e.g. 3 x IC90 value), harvested 16 to 24 h following treatment, washed twice with cold PBS and fixed in 70% ethanol at 4° C for at least 4 h. The fixed cells are centrifuged at 1500rpm for 4 minute at 4° C, washed twice with cold PBS containing 2% FBS, treated with 3 mg/ml ribonuclease (Sigma Chemical Co. Oakville, ON) and 50 µg/ml propidium iodide (PI) (Sigma Chemical Co.) for 30 minutes at 37° C. The fluorescence of the stained cells is measured using a FACScan flow cytometer and the Cell Quest program (Becton Dickinson, San Jose, CA). Data are evaluated using Modfit software (Verity software House, Topsham, ME) and the effects of the selected compounds on cell cycle are evaluated.
  • ii) Apoptosis assay
  • DNA fragmentation analysis is used to evaluate the apoptotic effects of test compounds. Briefly, cells are plated in six-well culture plates 24 hr prior to treatment. After incubation with the test compound, medium containing detached cells is transferred to 15 ml conical tubes while cells still attached to the plate are trypsinized and then added to the same tubes. After centrifugation, collected cells are washed with PBS and resuspended in 0.5 ml lysis buffer containing 50 mM Tris-HCl, pH 8.0, 1.0 M NaCl, 10 mM EDTA and 0.5% SDS. Cell lysates are transferred to microfuge tubes and proteinase K is added to a final concentration of 0.2 ml/ml and incubated overnight at 37° C. DNA is extracted by phenol:chloroform:isoamyl alcohol (24:24:1), dried and dissolved in 40 µM of 10 µM Tris-HCl (pH 8.0) and 0.1 mM EDTA. DNase-free RNase A is added to each sample for 30 min at 37° C and 12 µl of each sample are loaded onto a 2% agarose gel containing 0.5 µg/ml ethidium bromide and electrophoresed. DNA is visualized under UV illumination and the induction of apoptosis by the test compound is evaluated based on the generation of a nucleosomal-size DNA ladder.
  • iii) Anti-angiogenesis assay
  • Proliferation of new capillaries, i.e. angiogenesis or neovascularization, is critical for the transition of a small localized tumour to expand into a large malignant growth. The Matrigel Plug Assay (see, Passaniti et al., Lab. Invest. (1992) 67:519) is a simple method for assessing angiogenesis and the possible anti-angiogenic effect of selected compounds in mice. Briefly, liquid Matrigel (Becton Dickinson & Co., NJ) is injected subcutaneously near the abdominal midline or the dorsal flank of the animal using a 25-gauge needle. Growth factor-reduced Matrigel supplemented with 8.3 nM basic fibroblast growth factor (bFGF, Collaborative Biomedical Products, MA) stays in liquid form at 4 °C. bFGF is a proven and potent inducer of angiogenesis. When injected into a mouse (0.5 ml/mouse), Matrigel immediately forms a readily recoverable solid gel, which is removed at various times (not exceeding 10 days) to assess neo-vessel growth around and into the gel. Test compounds are administered according to appropriate doses and schedules. Typically at a 5-day point, mice are sacrificed, overlying skin is removed and the gels are cut out retaining the peritoneal lining for support. For quantitation of angiogenesis, two methods are employed: 1. haemoglobin content in the gel is measured using the Drabkin method (Drabkin and Austin, J. Biol Chem. (1932) 98:719) and Drabkin reagent kit 525 (Sigma, MO); 2. the number of blood vessels invading the Matrigel is determined by microscopic analysis after the gels are fixed, embedded in paraffin, sectioned and stained.
  • iv) Immunohistochemistry
  • The anti-cancer effects of test compounds can be evaluated in mouse xenograft models (as described above) by quantitating the effects of these compounds on tumour growth, differentiation, apoptosis and angiogenesis using immunohistochemical methods.
  • Tumour cell proliferation, angiogenesis and tumour immune infiltrates are delineated immunohistochemically using specific antibodies (Ki-67 for proliferation, CD31 for angiogenesis and NK1.1 for NK cells and F4/80 for macrophage). Apoptosis is delineated utilising the TUNEL assay (In Situ Cell Death Detection kit; Boehringer Mannheim, Laval, QC). Signal generation is accomplished by peroxidase catalyzed generation of enzyme product which is visualized microscopically. Tissue histology is determined after H&E staining of separate sections.
  • Briefly, tumour xenografts from treated mice are isolated, fixed and paraffin embedded individually in blocks and several 5 µm sections are cut for immunostaining and TUNEL assays. One additional section is obtained for H&E staining. For all immunohistochemical labeling, prior antigen retrieval is employed to improve detection. Typically, a 3-step amplification method is used to generate signals in immunohistochemistry that consists essentially of applying a biotinylated secondary antibody that recognizes the primary antibody, followed by avidin-peroxidase incubation. The final step is enzyme reaction in stable DAB solution. Immunohistochemical sections are counterstained with hematoxylin for tissue histology. To eliminate non-specific immunostaining with mouse monoclonal antibodies applied to mouse tissues, a specific blocking step is included in the procedure. Staining patterns are documented photographically, examined by at least two independent observers and quantitated by counting a pre-determined number of cells.

Claims (21)

  1. A compound of formula (VI), or a salt thereof, for use in the treatment of cancer:
    Figure imgb0086
    wherein:
    R4, R5, R6, R7, R8 and R9 are independently selected from hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, lower alkenyl, substituted lower alkenyl, lower alkynyl, substituted lower alkynyl, alkylalkenyl, alkylalkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro, cyano, or -S(O)0-2R wherein R is alkyl, substituted alkyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, or substituted heteroaryl;
    x is CR11;
    y is CR12 or N;
    z is CR13 or N;
    r is CR14 or N;
    x' is CR15;
    y' is CR16 or N;
    z' is CR17 or N;
    r' is CR18 or N;
    R10 is hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, or acyl; and
    R11, R12, R13, R14, R15, R16, R17, and R18 are independently hydrogen, halogen, hydroxyl, thiol, lower alkyl, substituted lower alkyl, alkenyl, alkenyl, alkylalkenyl, alkylalkynyl, alkoxy, alkylthio, acyl, aryloxy, amino, amido, carboxyl, aryl, substituted aryl, heterocycle, heteroaryl, substituted heterocycle, heteroalkyl, cycloalkyl, substituted cycloalkyl, alkylcycloalkyl, alkylcycloheteroalkyl, nitro or cyano,
    wherein:
    lower alkyl refers to a straight chain or branched alkyl group of 1 to 10 carbon atoms or a cyclic alkyl group of 3 to 10 carbon atoms;
    lower alkenyl refers to a straight chain or branched hydrocarbon of 2 to 10 carbon atoms or a cyclic hydrocarbon of 3 to 10 carbon atoms, having at least one carbon to carbon double bond;
    lower alkynyl refers to a straight chain or branched hydrocarbon of 2 to 10 carbons having at least one carbon to carbon triple bond.
  2. The compound for use according to claim 1, wherein R4 is hydrogen.
  3. The compound for use according to claim 1, wherein R10 is hydrogen.
  4. A compound selected from the group consisting of:
    Figure imgb0087
    Figure imgb0088
    Figure imgb0089
    Figure imgb0090
    Figure imgb0091
    Figure imgb0092
  5. A compound according to claim 4, wherein said compound is:
    Figure imgb0093
  6. A compound selected from:
    Figure imgb0094
    Figure imgb0095
    Figure imgb0096
    Figure imgb0097
    Figure imgb0098
    Figure imgb0099
    Figure imgb0100
    Figure imgb0101
    Figure imgb0102
    Figure imgb0103
    Figure imgb0104
    Figure imgb0105
    Figure imgb0106
    Figure imgb0107
    Figure imgb0108
    Figure imgb0109
    Figure imgb0110
    Figure imgb0111
    Figure imgb0112
    Figure imgb0113
    Figure imgb0114
    Figure imgb0115
    Figure imgb0116
    Figure imgb0117
    Figure imgb0118
    Figure imgb0119
    Figure imgb0120
    Figure imgb0121
    Figure imgb0122
    Figure imgb0123
    Figure imgb0124
    Figure imgb0125
    Figure imgb0126
    Figure imgb0127
    Figure imgb0128
    Figure imgb0129
    Figure imgb0130
    Figure imgb0131
  7. A pharmaceutical composition comprising the compound of any one of claims 4-6 and a pharmaceutically acceptable carrier.
  8. A compound according to any one of claims 4-6 for use in the treatment of cancer.
  9. The compound for use according to claim 1 or 8, wherein the cancer is a solid tumour.
  10. The compound for use according to claim 9, wherein the solid tumour is selected from the group consisting of cancers of the brain, breast, cervix, colon, head and neck, kidney, lung, ovary, pancreas, prostate, stomach, uterus, non-small cell lung cancer and colorectal cancer.
  11. The compound for use according to claim 1 or 8, wherein the cancer is leukemia.
  12. The compound for use according to claim 1 or 8, wherein the cancer is lymphoma.
  13. The compound for use according to claim 1 or 8, wherein the cancer is a refractory cancer.
  14. The compound for use according to claim 1 or 8, wherein the cancer is selected from the group consisting of Hodgkin's Disease, Non-Hodgkin's lymphoma, multiple myeloma, neuroblastoma, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small-cell lung tumours, primary brain tumours, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, gliomas, testicular cancer, thyroid cancer, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, mesothelioma and medulloblastoma.
  15. The compound for use according to any one of claims 1 and 8-14, wherein the compound is formulated for administration in combination with one or more anti-cancer agents(s).
  16. The compound for use according to any one of claims 1 and 8-14, wherein the compound is formulated for systemic administration.
  17. The compound for use according to claim 1 or 8, wherein the cancer is a stomach cancer.
  18. The compound for use according to claim 1 or 8, wherein the cancer is a multiple myeloma.
  19. The compound for use according to claim 11, wherein leukemia is adult T-cell leukemia.
  20. The compound for use according to claim 11, wherein the leukemia is acute or chronic.
  21. The compound for use according to claim 11 or claim 20, wherein the leukemia is myelogenous, lymphogenous, or monocytic.
EP04799154.2A 2003-11-14 2004-11-15 Aryl imidazoles and their use as anti-cancer agents Not-in-force EP1692113B1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US52027903P 2003-11-14 2003-11-14
US59950904P 2004-08-06 2004-08-06
PCT/IB2004/052433 WO2005047266A1 (en) 2003-11-14 2004-11-15 Aryl imidazoles and their use as anti-cancer agents

Publications (2)

Publication Number Publication Date
EP1692113A1 EP1692113A1 (en) 2006-08-23
EP1692113B1 true EP1692113B1 (en) 2017-09-27

Family

ID=34594988

Family Applications (1)

Application Number Title Priority Date Filing Date
EP04799154.2A Not-in-force EP1692113B1 (en) 2003-11-14 2004-11-15 Aryl imidazoles and their use as anti-cancer agents

Country Status (7)

Country Link
US (2) US8969372B2 (en)
EP (1) EP1692113B1 (en)
JP (1) JP5095216B2 (en)
AU (1) AU2004289539C1 (en)
CA (1) CA2545942C (en)
DK (1) DK1692113T3 (en)
WO (1) WO2005047266A1 (en)

Families Citing this family (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2497116T3 (en) 2002-08-19 2014-09-22 Lorus Therapeutics Inc. 2,4,5-trisubstituted imidazoles and their use as antimicrobial agents
TWI372050B (en) 2003-07-03 2012-09-11 Astex Therapeutics Ltd (morpholin-4-ylmethyl-1h-benzimidazol-2-yl)-1h-pyrazoles
AU2004289539C1 (en) * 2003-11-14 2012-06-07 Lorus Therapeutics Inc. Aryl imidazoles and their use as anti-cancer agents
US7713973B2 (en) 2004-10-15 2010-05-11 Takeda Pharmaceutical Company Limited Kinase inhibitors
US7442716B2 (en) 2004-12-17 2008-10-28 Merck Frosst Canada Ltd. 2-(phenyl or heterocyclic)-1H-phenantrho[9,10-d]imidazoles as mPGES-1 inhibitors
RU2416610C2 (en) 2004-12-30 2011-04-20 Астекс Терапьютикс Лимитед Pharmaceutical compounds
JP5334575B2 (en) * 2005-05-25 2013-11-06 ローラス セラピューティクス インコーポレーテッド 2-Indolylimidazo [4,5-d] phenanthroline derivatives and their use in cancer treatment
US8148392B2 (en) 2005-05-25 2012-04-03 Lorus Therapeutics Inc. 2-indolyl imidazo [4,5-d] phenanthroline derivatives and their use in the treatment of cancer
SG162803A1 (en) * 2005-06-27 2010-07-29 Exelixis Inc Imidazole based lxr modulators
ES2525217T3 (en) * 2005-06-27 2014-12-19 Exelixis Patent Company Llc LXR modulators based on imidazole
US8119655B2 (en) 2005-10-07 2012-02-21 Takeda Pharmaceutical Company Limited Kinase inhibitors
US8399442B2 (en) 2005-12-30 2013-03-19 Astex Therapeutics Limited Pharmaceutical compounds
US20090192158A1 (en) * 2006-05-02 2009-07-30 Stacia Kargman Methods for Treating or Preventing Neoplasias
EP2049119A2 (en) 2006-06-29 2009-04-22 Astex Therapeutics Limited Pharmaceutical combinations of 1-cyclopropyl-3-[3-(5-morphoolin-4-ylmethyl-1h-benzoimidazol-2-yl)-1h-1-pyrazol-4-yl]-urea
US20080090834A1 (en) * 2006-07-06 2008-04-17 Pfizer Inc Selective azole pde10a inhibitor compounds
JP2010505962A (en) 2006-10-09 2010-02-25 武田薬品工業株式会社 Kinase inhibitor
JP5399262B2 (en) 2006-12-08 2014-01-29 エグゼリクシス パテント カンパニー エルエルシー LXR and FXR modulators
WO2008105565A1 (en) * 2007-02-26 2008-09-04 Industry-Academic Cooperation Foundation, Yonsei University Imidazole derivatives that induce apoptosis and their therapeutic uses
CN101130539B (en) * 2007-09-26 2012-05-30 浙江大学 Indole-substituteing imidazoline-2-ketones derivant, preparing method and uses of the same
US8865732B2 (en) 2008-03-21 2014-10-21 Novartis Ag Heterocyclic compounds and uses thereof
JP5384611B2 (en) 2008-03-21 2014-01-08 ノバルティス アーゲー Novel heterocyclic compounds and their use
MX2011003239A (en) 2008-09-26 2011-04-28 Merck Sharp & Dohme Novel cyclic benzimidazole derivatives useful anti-diabetic agents.
WO2010047982A1 (en) 2008-10-22 2010-04-29 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
CA2741672A1 (en) 2008-10-31 2010-05-06 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
WO2010102393A1 (en) * 2009-03-12 2010-09-16 Lorus Therapeutics Inc. 2-indolyl imidazo [4,5-d]phenanthroline derivatives and their use to inhibit angiogenesis
PE20120797A1 (en) * 2009-05-28 2012-07-08 Exelisis Patent Company Llc DERIVATIVES OF 1 - [(3'-FLUORO-4'-HYDROXIMETHYL-5'-METHYLSULFONYL) BIPHENYL-4-IL] -2-BENZYL-4 - [(1-HYDROXY-1-METHYL) ETHYL] -1H-IMIDAZOLE AS LXR MODULATORS
DK2987487T3 (en) 2009-08-10 2020-12-07 Samumed Llc INDAZOLINE INHIBITORS OF THE WNT SIGNAL ROAD AND ITS THERAPEUTIC USES
JO3002B1 (en) 2009-08-28 2016-09-05 Irm Llc Compounds and compositions as protein kinase inhibitors
CN101735217B (en) * 2009-12-15 2012-09-26 广东药学院 Application of imidazole [4,5-f][1,10] phenanthroline and derivative thereof to preparation of antineoplastic drug
DK3001903T3 (en) 2009-12-21 2017-12-18 Samumed Llc 1H-PYRAZOLO [3,4 -?] PYRIDINES AND THERAPEUTIC APPLICATIONS THEREOF
WO2011106273A1 (en) 2010-02-25 2011-09-01 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
RU2635662C2 (en) 2010-09-10 2017-11-15 Сионоги Энд Ко., Лтд. Imidzol derivative condensed heterocycle having active ampk effect
DE102011009961A1 (en) 2011-02-01 2012-08-02 Merck Patent Gmbh 7-azaindole derivatives
AU2012308570B2 (en) 2011-09-14 2016-11-10 Samumed, Llc Indazole-3-carboxamides and their use as Wnt/b-catenin signaling pathway inhibitors
CA2855243C (en) 2011-11-11 2020-04-14 Novartis Ag Method of treating a proliferative disease
PH12017500997A1 (en) 2012-04-04 2018-02-19 Samumed Llc Indazole inhibitors of the wnt signal pathway and therapeutic uses thereof
DK2770994T3 (en) 2012-05-04 2019-11-11 Samumed Llc 1H-PYRAZOLO [3,4-B] PYRIDINES AND THERAPEUTIC APPLICATIONS THEREOF
CN105120862A (en) 2013-01-08 2015-12-02 萨穆梅德有限公司 3-(benzoimidazol-2-yl)-indazole inhibitors of the wnt signaling pathway and therapeutic uses thereof
US9242969B2 (en) 2013-03-14 2016-01-26 Novartis Ag Biaryl amide compounds as kinase inhibitors
JP2016516082A (en) * 2013-03-20 2016-06-02 アプトース バイオサイエンシーズ, インコーポレイテッド 2-Substituted imidazo [4,5-D] phenanthroline derivatives and their use in the treatment of cancer
DK3052102T3 (en) 2013-10-04 2020-03-09 Aptose Biosciences Inc CANCER TREATMENT COMPOSITIONS
AU2015240465B2 (en) 2014-04-04 2020-02-27 Del Mar Pharmaceuticals Use of dianhydrogalactitol and analogs or derivatives thereof to treat non-small-cell carcinoma of the lung and ovarian cancer
WO2016040182A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 2-(1h-indazol-3-yl)-1h-imidazo[4,5-c]pyridine and therapeutic uses thereof
WO2016040185A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 2-(1h-indazol-3-yl)-3h-imidazo[4,5-b]pyridine and therapeutic uses thereof
WO2016040184A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040180A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-benzo[d]imidazol-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040181A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
WO2016040193A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(1h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
WO2016040190A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[3,4-b]pyridine and therapeutic uses thereof
WO2016040188A1 (en) 2014-09-08 2016-03-17 Samumed, Llc 3-(3h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridine and therapeutic uses thereof
UY36294A (en) 2014-09-12 2016-04-29 Novartis Ag COMPOUNDS AND COMPOSITIONS AS QUINASA INHIBITORS
WO2016086022A2 (en) * 2014-11-26 2016-06-02 Channel Therapeutics, Inc. Heteroaromatic imidazolyl compounds and methods for treating cancer
CA3030582A1 (en) 2015-07-22 2017-01-26 The Royal Institution For The Advancement Of Learning/Mcgill University Compounds and uses thereof in the treatment of cancers and other medical conditions
US10226453B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-indol-2-yl)-1H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
WO2017024004A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10463651B2 (en) 2015-08-03 2019-11-05 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-indazoles and therapeutic uses thereof
WO2017024015A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(3h-imidazo[4,5-b]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10206908B2 (en) 2015-08-03 2019-02-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
WO2017024021A1 (en) 2015-08-03 2017-02-09 Samumed, Llc 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-indazoles and therapeutic uses thereof
WO2017023980A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-pyrrolo[2,3-b]pyridin-2-yl)-1h-pyrazolo[3,4-c]pyridines and therapeutic uses thereof
WO2017024026A1 (en) 2015-08-03 2017-02-09 Samumed, Llc 3-(1h-indol-2-yl)-1h-pyrazolo[3,4-c]pyridines and therapeutic uses thereof
US10392383B2 (en) 2015-08-03 2019-08-27 Samumed, Llc 3-(1H-benzo[d]imidazol-2-yl)-1H-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10383861B2 (en) 2015-08-03 2019-08-20 Sammumed, LLC 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1H-pyrazolo[3,4-C]pyridines and therapeutic uses thereof
US10226448B2 (en) 2015-08-03 2019-03-12 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-yl)-1H-pyrazolo[3,4-B]pyridines and therapeutic uses thereof
US10285983B2 (en) 2015-08-03 2019-05-14 Samumed, Llc 3-(1H-pyrrolo[2,3-B]pyridin-2-yl)-1H-pyrazolo[3,4-B] pyridines and therapeutic uses thereof
US10604512B2 (en) 2015-08-03 2020-03-31 Samumed, Llc 3-(1H-indol-2-yl)-1H-indazoles and therapeutic uses thereof
US10231956B2 (en) 2015-08-03 2019-03-19 Samumed, Llc 3-(1H-pyrrolo[3,2-C]pyridin-2-YL)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
WO2017023972A1 (en) 2015-08-03 2017-02-09 Samumed, Llc. 3-(1h-imidazo[4,5-c]pyridin-2-yl)-1h-pyrazolo[4,3-b]pyridines and therapeutic uses thereof
US10188634B2 (en) 2015-08-03 2019-01-29 Samumed, Llc 3-(3H-imidazo[4,5-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
US10519169B2 (en) 2015-08-03 2019-12-31 Samumed, Llc 3-(1H-pyrrolo[2,3-C]pyridin-2-yl)-1 H-pyrazolo[4,3-B]pyridines and therapeutic uses thereof
CA3004297A1 (en) 2015-11-06 2017-05-11 Samumed, Llc 2-(1h-indazol-3-yl)-3h-imidazo[4,5-c]pyridines and their anti-inflammatory uses thereof
PL3464285T3 (en) 2016-06-01 2023-02-06 Biosplice Therapeutics, Inc. Process for preparing n-(5-(3-(7-(3-fluorophenyl)-3h-imidazo[4,5-c]pyridin-2-yl)-1h-indazol-5-yl)pyridin-3-yl)-3-methylbutanamide
CN106220612A (en) * 2016-08-10 2016-12-14 大连理工大学 One class is containing phenanthro-[9,10 d] imidazole derivative of indole and synthesis thereof and application
JP2019535672A (en) 2016-10-21 2019-12-12 サミュメッド リミテッド ライアビリティ カンパニー Methods of using indazole-3-carboxamides and their use as inhibitors of WNT / Β-catenin signaling pathway
MA46696A (en) 2016-11-07 2019-09-11 Samumed Llc READY-TO-USE SINGLE-DOSE INJECTABLE FORMULATIONS
KR102070879B1 (en) * 2017-10-17 2020-01-29 성균관대학교산학협력단 Pharmaceutical Composition for Preventing or Treating cancer comprising MNPⅡ as Active Ingredients
KR20200096914A (en) 2017-10-30 2020-08-14 압토스 바이오사이언시스 인코포레이티드 Aryl imidazole for cancer treatment
GB201718285D0 (en) 2017-11-03 2017-12-20 Discuva Ltd Antibacterial Compounds
WO2022249192A1 (en) * 2021-05-27 2022-12-01 Ramot At Tel-Aviv University Ltd. Broad-spectrum metastasis suppressing compounds and therapeutic uses thereof in human tumors

Family Cites Families (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE585507A (en) * 1960-03-31
US3297710A (en) * 1963-04-09 1967-01-10 Du Pont Oxoarylidene-imidazoles
US3714181A (en) * 1970-12-31 1973-01-30 American Cyanamid Co 2-aryl-5,10-diphenylphenanthro(9,10-d)azoles
US4089747A (en) * 1976-08-09 1978-05-16 Eastman Kodak Company Compositions for the detection of hydrogen peroxide
US4535058A (en) 1982-10-01 1985-08-13 Massachusetts Institute Of Technology Characterization of oncogenes and assays based thereon
DE3141063A1 (en) * 1981-10-13 1983-04-28 Schering Ag, 1000 Berlin Und 4619 Bergkamen NEW IMIDAZOLE DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL PREPARATIONS CONTAINING THEM
US4423046A (en) * 1982-04-05 1983-12-27 Sterling Drug Inc. Antibacterial and antiprotozoal 1-methyl-5-nitro-2-(2-phenylvinyl)imidazoles
DE3422175A1 (en) 1984-06-04 1985-12-19 Julius Dr. 8000 München Kern Process for adding ammonia to exhaust gases or air containing acidic pollutants
JPS614960A (en) 1984-06-19 1986-01-10 Fuji Photo Film Co Ltd Analyzing reagent, analyzing method and multilayered chemical analyzing element
US4762706A (en) 1984-10-17 1988-08-09 Cetus Corporation Peptide antibodies and their use in detecting oncogene products
US5443956A (en) 1985-01-29 1995-08-22 Oncogene Science, Inc. Detection, quantitation and classification of RAS proteins in body fluids and tissues
US5081230A (en) 1987-07-08 1992-01-14 E. I. Dupont Denemours And Company Monoclonal antibodies reactive with normal and oncogenic forms of the ras p21 protein
US4720459A (en) 1985-02-14 1988-01-19 Medical College Of Wisconsin Research Foundation, Inc. Myelomas for producing human/human hybridomas
US4758421A (en) 1985-03-15 1988-07-19 The Board Of Trustees Of The Leland Stanford Junior University Bleomycin conjugates and method
IT1214597B (en) * 1985-04-15 1990-01-18 Rorer Italiana Spa DERIVATIVES OF IMIDAZOLE ANTIMICOTIC ADAPTITY, PROCEDURE AND INTERMEDIATES FOR THEIR PREPARATION AND COMPOSITIONS CONTAINING THEM.
US5023252A (en) * 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
EP0248086B1 (en) * 1985-12-12 1993-03-17 Ube Industries, Ltd. Imidazole derivatives, bactericides containing them, and process for their preparation
US5024935A (en) * 1987-12-18 1991-06-18 Eastman Kodak Company Dye-providing composition, diagnostic test kit and their use in method for ligand determination using peroxidase labeled-receptor
US5047318A (en) * 1988-06-13 1991-09-10 Eastman Kodak Company Imidazole leuco dye composition containing 4'-hydroxyacetanilide, diagnostic kit and method using same
US5011472A (en) * 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
US5501959A (en) 1989-01-17 1996-03-26 Alamar Biosciences Laboratory, Inc. Antibiotic and cytotoxic drug susceptibility assays using resazurin and poising agents
US5514550A (en) * 1989-02-03 1996-05-07 Johnson & Johnson Clinical Diagnostics, Inc. Nucleic acid test article and its use to detect a predetermined nucleic acid
US5441716A (en) 1989-03-08 1995-08-15 Rocky Research Method and apparatus for achieving high reaction rates
EP0462226B1 (en) 1989-03-08 1995-05-10 Rocky Research Method and apparatus for achieving high reaction rates in solid-gas reactor systems
JPH02258017A (en) 1989-03-31 1990-10-18 Babcock Hitachi Kk Denitrifying apparatus for waste gas using solid reducing agent
US4970226A (en) * 1989-10-03 1990-11-13 Harbor Branch Oceanographic Institution, Inc. Bis-indole imidazole compounds which are useful antitumor and antimicrobial agents
US5161389A (en) 1990-11-13 1992-11-10 Rocky Research Appliance for rapid sorption cooling and freezing
US5916891A (en) * 1992-01-13 1999-06-29 Smithkline Beecham Corporation Pyrimidinyl imidazoles
MX9300141A (en) 1992-01-13 1994-07-29 Smithkline Beecham Corp NOVEL IMIDAZOLE COMPOUNDS, PROCEDURE FOR THE PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING IT.
US5656644A (en) * 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
WO1995003297A1 (en) 1993-07-21 1995-02-02 Smithkline Beecham Corporation Imidazoles for treating cytokine mediated disease
US5496702A (en) * 1994-09-01 1996-03-05 Johnson & Johnson Clinical Diagnostics, Inc. Immunoassay elements having stable leuco dye coatings
EP0797575B1 (en) 1994-12-13 2003-10-15 F. Hoffmann-La Roche Ag Imidazole derivatives as protein kinase inhibitors in particular egf-r tyrosine kinase
TW424172B (en) * 1995-04-19 2001-03-01 Hitachi Chemical Co Ltd Photosensitive resin composition and photosensitive element using the same
US5700826A (en) * 1995-06-07 1997-12-23 Ontogen Corporation 1,2,4,5-tetra substituted imidazoles as modulators of multi-drug resistance
US5753687A (en) * 1995-06-19 1998-05-19 Ontogen Corporation Modulators of proteins with phosphotryrosine recognition units
JP2000504023A (en) * 1996-04-03 2000-04-04 メルク エンド カンパニー インコーポレーテッド Cancer treatment methods
US5693589A (en) * 1996-05-08 1997-12-02 Eastman Kodak Company Thermal imaging recording element
EP0812829A1 (en) * 1996-06-14 1997-12-17 Ontogen Corporation Substituted imidazoles as modulators of multi-drug resistance
IT240917Y1 (en) 1996-10-01 2001-04-11 Stefcom Spa SHOE WITH UPPER HAVING PORTION FOR BREATHING THE INTERIOR OF THE SHOE
WO1998027108A2 (en) 1996-12-16 1998-06-25 Fujisawa Pharmaceutical Co., Ltd. New amide compounds and their use as nitric oxide synthase inhibitors
AU740425B2 (en) * 1996-12-16 2001-11-01 Ontogen Corporation Modulators of proteins with phosphotyrosine recognition units
US5945418A (en) * 1996-12-18 1999-08-31 Vertex Pharmaceuticals Incorporated Inhibitors of p38
US5809775A (en) 1997-04-02 1998-09-22 Clean Diesel Technologies, Inc. Reducing NOx emissions from an engine by selective catalytic reduction utilizing solid reagents
EP1028954B1 (en) * 1997-04-24 2003-07-02 Ortho-McNeil Pharmaceutical, Inc. Substituted imidazoles useful in the treatment of inflammatory diseases
JPH10319584A (en) * 1997-05-21 1998-12-04 Brother Ind Ltd Photosensitive recording material using microcapsules
DE19728343C5 (en) 1997-07-03 2013-02-21 Robert Bosch Gmbh Process and apparatus for selective catalytic NOx reduction
PL337887A1 (en) 1997-07-03 2000-09-11 Neurogen Corp Some diarylimidazole derivatives, novel class of specific npy ligands
US5840721A (en) 1997-07-09 1998-11-24 Ontogen Corporation Imidazole derivatives as MDR modulators
WO1999007701A1 (en) * 1997-08-05 1999-02-18 Sugen, Inc. Tricyclic quinoxaline derivatives as protein tyrosine kinase inhibitors
GB9717804D0 (en) * 1997-08-22 1997-10-29 Zeneca Ltd Chemical compounds
JPH11199582A (en) 1998-01-05 1999-07-27 Sagami Chem Res Center 4(5)-(3-indolyl) imidazole derivative
US5915891A (en) 1998-02-17 1999-06-29 Fridman; Yevgeny Z. Drill guide and method for installing a door lock
JP2002506855A (en) 1998-03-18 2002-03-05 ウェイク フォレスト ユニヴァーシティ Cytotoxic metal chelating agents, their preparation and use
US6288212B1 (en) 1998-08-28 2001-09-11 The University Of British Columbia Anti-endotoxic, antimicrobial cationic peptides and methods of use therefor
AU2164300A (en) 1998-12-04 2000-06-26 N.V. Organon Substituted thiazoles for treatment of human diseases involving modulators of p-, l- and e- selectin
JP2000273088A (en) 1999-01-18 2000-10-03 Nippon Soda Co Ltd Triphenylimidazole
JP2000281588A (en) * 1999-03-30 2000-10-10 Sankyo Co Ltd Prophylactic or therapeutic agent for cancer and method for screening same
DE19920936A1 (en) 1999-05-07 2000-11-09 Basf Ag Heterocyclically substituted benzimidazoles, their preparation and use
AU4640600A (en) 1999-05-11 2000-11-21 Eli Lilly And Company Amyloid precursor protein protease and related nucleic acid compounds
US7364868B2 (en) 1999-05-19 2008-04-29 The Uab Research Foundation Krüppel-like transcriptional factor KLF4/GKLF and uses thereof
DE19925631A1 (en) 1999-06-04 2000-12-07 Georg Gros Coating process and coating mixture
AU5886500A (en) * 1999-06-23 2001-01-09 Sepracor, Inc. Indolyl-benzimidazole antibacterials, and methods of use thereof
US6266955B1 (en) 1999-08-20 2001-07-31 Caterpillar Inc. Diagnostic system for an emissions control on an engine
CN1087744C (en) 1999-09-24 2002-07-17 中国科学院化学研究所 Ruthenium (II) polypyridine match and its preparing process
CN1088713C (en) 1999-09-24 2002-08-07 中国科学院化学研究所 Ruthenium (II) polypyridine match for two-stage pH sensing and its preparing process
ATE426335T1 (en) * 1999-10-08 2009-04-15 Taiji Biomedical Inc METHOD FOR IMPROVING CHEMOTHERAPY
US7115645B2 (en) * 2000-01-14 2006-10-03 Schering Aktiengesellschaft 1,2 diarylbenzimidazoles and their pharmaceutical use
DE60140865D1 (en) 2000-03-27 2010-02-04 Univ Jefferson COMPOSITIONS AND METHODS OF IDENTIFYING CANCER CELLS
KR20030030027A (en) 2000-09-21 2003-04-16 스미스클라인비이참피이엘시이 Imidazole Derivatives As Raf Kinase Inhibiotrs
ATE510539T1 (en) 2000-10-13 2011-06-15 Nanotherapeutics Inc MODIFIED PRODRUG FORMS OF AP
US20070004713A1 (en) 2000-12-07 2007-01-04 Bernard Barlaam Therapeutic benimidazole compounds
BR0207957A (en) * 2001-03-09 2004-02-25 Pfizer Prod Inc Benzimidazole Anti-Inflammatory Compounds
JP2002275458A (en) 2001-03-21 2002-09-25 Fukuoka Prefecture Photochromic amorphous material and high density optical recording medium
KR100863659B1 (en) 2001-04-16 2008-10-15 미쓰비시 타나베 파마 코퍼레이션 Nitrogen-Containing Heterocyclic Compound and Pharmaceutical Composition Thereof
US6916440B2 (en) 2001-05-31 2005-07-12 3M Innovative Properties Company Processes and apparatus for making transversely drawn films with substantially uniaxial character
JP3759882B2 (en) 2001-06-01 2006-03-29 株式会社日立製作所 Pump device
DE10134775A1 (en) 2001-07-06 2003-01-30 Schering Ag 1-alkyl-2.aryl-benzimidazole derivatives, their use for the manufacture of medicaments and pharmaceutical preparations containing these derivatives
US7695926B2 (en) 2001-07-10 2010-04-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for detecting receptor-ligand interactions in single cells
US6852727B2 (en) * 2001-08-01 2005-02-08 Merck & Co., Inc. Benzimisazo[4,5-f]isoquinolinone derivatives
CA2464000C (en) * 2001-10-19 2011-04-19 Ortho-Mcneil Pharmaceutical, Inc. 2-phenyl benzimidazoles and imidazo-[4,5]-pyridines as cdsi/chk2-inhibitors and adjuvants to chemotherapy or radiation therapy in the treatment of cancer
JP2005517007A (en) 2002-02-07 2005-06-09 アクシス・ファーマスーティカルズ Novel bicyclic hydroxamates as histone deacetylase inhibitors
US6790852B2 (en) * 2002-04-18 2004-09-14 Hoffmann-La Roche Inc. 2-(2,6-dichlorophenyl)-diarylimidazoles
EP1428831A4 (en) 2002-06-19 2006-09-20 Kurita Water Ind Ltd Method of storing hydrogen, hydrogen inclusion compound and process for producing the same
EP1532118A2 (en) 2002-07-05 2005-05-25 Axxima Pharmaceuticals Aktiengesellschaft Imidazole compounds for the treatment of hepatitis c virus infections
ES2497116T3 (en) 2002-08-19 2014-09-22 Lorus Therapeutics Inc. 2,4,5-trisubstituted imidazoles and their use as antimicrobial agents
DE10251472A1 (en) 2002-11-06 2004-05-19 Robert Bosch Gmbh Reduction of automotive nitrous oxide emissions during cold start comprises supplementary injection of ammonia from holding reservoir
WO2004071464A2 (en) 2003-02-12 2004-08-26 Johns Hopkins University School Of Medicine Diagnostic application of differentially-expressed genes in lympho-hematopoietic stem cells
CA2425797C (en) * 2003-04-17 2013-10-15 Xerox Corporation Organic light emitting devices
US7989089B2 (en) * 2003-04-17 2011-08-02 Lg Display Co., Ltd. Organic luminescent compounds and methods of making and using same
DE10323591A1 (en) 2003-05-16 2004-12-02 E.G.O. Elektro-Gerätebau GmbH Device for producing a gas from a sublimation material, preferably ammonia from ammonium carbamate, comprises a heating arrangement assigned to sublimation material in a container
AU2004289539C1 (en) 2003-11-14 2012-06-07 Lorus Therapeutics Inc. Aryl imidazoles and their use as anti-cancer agents
US20050195793A1 (en) 2004-03-05 2005-09-08 Lockheed Martin Corporation System for locally synchronizing a group of mobile devices
US20050282285A1 (en) 2004-06-21 2005-12-22 Eaton Corporation Strategy for controlling NOx emissions and ammonia slip in an SCR system using a nonselective NOx/NH3
KR20070057143A (en) 2004-08-03 2007-06-04 아미넥스 에이/에스 A solid ammonia storage and delivery material
RU2395335C2 (en) 2005-02-03 2010-07-27 Амминекс А/С Storage of high-content ammonia
US8148392B2 (en) * 2005-05-25 2012-04-03 Lorus Therapeutics Inc. 2-indolyl imidazo [4,5-d] phenanthroline derivatives and their use in the treatment of cancer
JP2008546968A (en) 2005-06-29 2008-12-25 アムミネクス・アー/エス Method and apparatus for the safe and controlled delivery of ammonia from a solid ammonia storage medium
KR100694181B1 (en) * 2005-11-25 2007-03-12 연세대학교 산학협력단 Compounds inducing differentiation of myoblasts or muscle fibers into neuron cells, pharmaceutical composition including said compounds, a method for inducing neuron differentiation and a screening method for identifying additional compound useful for inducing neuron differentiation
DK2063907T3 (en) 2006-09-07 2017-11-13 Arthur E Frankel METHODS AND COMPOSITIONS BASED ON DIFTERY TOXIN-INTERLEUKIN-3 CONJUGATES
WO2008125883A1 (en) 2007-04-16 2008-10-23 Cancer Research Technology Limited Cancer markers for prognosis and screening of anti-cancer agents
US8507498B2 (en) 2007-04-24 2013-08-13 Ingenium Pharmaceuticals Gmbh 4, 6-disubstituted aminopyrimidine derivatives as inhibitors of protein kinases
WO2009058394A1 (en) 2007-11-01 2009-05-07 Celgene Corporation Cytidine analogs for treatment of myelodysplastic syndromes
WO2009151503A2 (en) 2008-04-10 2009-12-17 University Of Florida Research Foundation, Inc. Compositions and methods for the treatment of a neoplasia
CA2695590C (en) 2008-06-27 2018-01-09 Kyoto University Method of efficiently establishing induced pluripotent stem cells
US8399206B2 (en) 2008-07-10 2013-03-19 Nodality, Inc. Methods for diagnosis, prognosis and methods of treatment
WO2010102393A1 (en) 2009-03-12 2010-09-16 Lorus Therapeutics Inc. 2-indolyl imidazo [4,5-d]phenanthroline derivatives and their use to inhibit angiogenesis
EP2239580B1 (en) 2009-04-09 2013-03-27 Lga Biotecnologie Srl Determination of 5-ASA efficacy in CRC prevention and/or treatment by gene expression analysis
EP2417984B1 (en) 2009-04-10 2016-03-30 Kyowa Hakko Kirin Co., Ltd. Method for treatment of blood tumor using anti-tim-3 antibody
EP2491116A4 (en) 2009-10-22 2013-12-11 Univ Jefferson Cell-based anti-cancer compositions and methods of making and using the same
US8524240B2 (en) 2009-10-26 2013-09-03 Djordje Atanackovic Diagnosis and therapy of hematological malignancies
US20130011411A1 (en) 2010-01-06 2013-01-10 Pestell Richard G Methods and compositions for the diagnosis, prognosis, and treatment of cancer
KR101317513B1 (en) 2010-05-17 2013-10-15 가톨릭대학교 산학협력단 Composition, kit and method for diagnosing colorectal cancer or ovarian cancer
WO2012006032A2 (en) 2010-06-28 2012-01-12 Threshold Pharmaceuticals, Inc. Treatment of blood cancer
US20120172244A1 (en) 2010-12-20 2012-07-05 Steven Buechler Biomarkers and uses thereof in prognosis and treatment strategies for right-side colon cancer disease and left-side colon cancer disease
AU2012347460B2 (en) 2011-12-09 2017-05-25 Adaptive Biotechnologies Corporation Diagnosis of lymphoid malignancies and minimal residual disease detection
TW201343920A (en) 2012-03-29 2013-11-01 Nat Health Research Institutes Molecular markers for prognostically predicting prostate cancer, method and kit thereof
DK3052102T3 (en) 2013-10-04 2020-03-09 Aptose Biosciences Inc CANCER TREATMENT COMPOSITIONS

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
GAO D ET AL: "Synthesis and electroluminescence properties of an organic europium complex", JOURNAL OF ALLOYS AND COMPOUNDS, ELSEVIER SEQUOIA, LAUSANNE, CH, vol. 358, no. 1-2, 25 August 2003 (2003-08-25), pages 188 - 192, XP004448427, ISSN: 0925-8388, DOI: 10.1016/S0925-8388(03)00048-3 *
HONG XU ET AL: "Effects of the ancillary ligands of polypyridyl ruthenium(ii) complexes on the DNA-binding behaviors", NEW JOURNAL OF CHEMISTRY, vol. 27, no. 8, 1 January 2003 (2003-01-01), pages 1255, XP055083557, ISSN: 1144-0546, DOI: 10.1039/b212826h *
MIN GUAN ET AL: "Bright red light-emitting electroluminescence devices based on a functionalized europium complex", NEW JOURNAL OF CHEMISTRY, vol. 27, no. 12, 1 January 2003 (2003-01-01), pages 1731, XP055083545, ISSN: 1144-0546, DOI: 10.1039/b305361j *
XU H ET AL: "Synthesis and spectroscopic RNA binding studies of [Ru(phen)2MHPIP]<2+>", INORGANIC CHEMISTRY COMMUNICATIONS, ELSEVIER, AMSTERDAM, NL, vol. 6, no. 6, 1 June 2003 (2003-06-01), pages 766 - 768, XP027113034, ISSN: 1387-7003, [retrieved on 20030601] *
YA XIONG ET AL: "Interaction of polypyridyl ruthenium(II) complexes containing non-planar ligands with DNA", JOURNAL OF THE CHEMICAL SOCIETY, DALTON TRANSACTIONS, no. 1, 1 January 1999 (1999-01-01), pages 19 - 24, XP055083547, ISSN: 0300-9246, DOI: 10.1039/a806170j *

Also Published As

Publication number Publication date
US20080262015A9 (en) 2008-10-23
US10080739B2 (en) 2018-09-25
AU2004289539B2 (en) 2011-11-24
WO2005047266A1 (en) 2005-05-26
CA2545942A1 (en) 2005-05-26
US20070123553A1 (en) 2007-05-31
EP1692113A1 (en) 2006-08-23
WO2005047266A9 (en) 2008-04-17
CA2545942C (en) 2012-07-10
US20150374669A1 (en) 2015-12-31
AU2004289539A1 (en) 2005-05-26
JP2007511504A (en) 2007-05-10
US8969372B2 (en) 2015-03-03
JP5095216B2 (en) 2012-12-12
AU2004289539C1 (en) 2012-06-07
DK1692113T3 (en) 2018-01-08

Similar Documents

Publication Publication Date Title
EP1692113B1 (en) Aryl imidazoles and their use as anti-cancer agents
DE60120494T2 (en) SUBSTITUTES TRIAZOLDIAMINE DERIVATIVES AND THEIR USE AS KINASE INHIBITORS
EP2086525B1 (en) Method of radio-sensitizing tumors using a radio-sensitizing agent
EA015779B1 (en) Compounds for inhibiting mitotic progression
BR112013028095B1 (en) Use of csf-1r inhibitors for the treatment of brain tumors
CA3138197A1 (en) Anti-cancer nuclear hormone receptor-targeting compounds
WO2007067752A2 (en) Certain compositions and methods of treatment
EP3062790A1 (en) Pharmaceutical combinations for the treatment of cancer
Bharti et al. Pathways linked to cancer chemoresistance and their targeting by nutraceuticals
CN107207510B (en) Conjoint therapy
KR20070064414A (en) Use of chk1 inhibitors to control cell proliferation
US20180169123A1 (en) Combination therapy with a flavagline and 2-deoxyglucose
EP3180004B1 (en) Cancer therapeutics
IL293810A (en) Use of atr inhibitors in combination with parp inhibitors
KR102189562B1 (en) Acyl-hydrazone and oxadiazole compounds, pharmaceutical compositions containing the same and uses thereof
MXPA05009708A (en) Method of treating cancer with azaspirane compositions.
TWI607752B (en) Use of a composition containing 4-acetyl-antroquinonol b for preparing pharmaceutical compositions for inhibiting growth of ovarian cancer cells
WO2022261643A1 (en) Treating cancers with a cyclin-dependent kinase inhibitor
Visa et al. Hedhehog as a New Paradigm in Cancer Treatment
KR20100004681A (en) Use in anti-cancer agent of 5,4&#39;-dimethoxyflavone showing regulatory effect on p21

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20060606

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LU MC NL PL PT RO SE SI SK TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1094798

Country of ref document: HK

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GENESENSE TECHNOLOGIES INC.

RIN1 Information on inventor provided before grant (corrected)

Inventor name: YOUNG, AIPING, H.

Inventor name: HUESCA, MARIO

Inventor name: LEE, YOON

Inventor name: LOCK LISA

Inventor name: AL-QAWASMEH, RAED

17Q First examination report despatched

Effective date: 20071024

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/4178 20060101ALI20080509BHEP

Ipc: C07D 495/04 20060101ALI20080509BHEP

Ipc: C07D 235/02 20060101ALI20080509BHEP

Ipc: C07D 403/04 20060101ALI20080509BHEP

Ipc: C07D 403/14 20060101ALI20080509BHEP

Ipc: C07D 233/54 20060101AFI20080509BHEP

Ipc: A61K 31/4188 20060101ALI20080509BHEP

Ipc: C07D 405/14 20060101ALI20080509BHEP

Ipc: C07D 409/14 20060101ALI20080509BHEP

Ipc: A61K 31/4164 20060101ALI20080509BHEP

Ipc: C07D 471/14 20060101ALI20080509BHEP

Ipc: C07D 401/04 20060101ALI20080509BHEP

Ipc: A61K 31/4184 20060101ALI20080509BHEP

Ipc: C07D 401/14 20060101ALI20080509BHEP

Ipc: C07D 471/04 20060101ALI20080509BHEP

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: LORUS THERAPEUTICS INC.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20170418

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LU MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602004051855

Country of ref document: DE

Owner name: APTOSE BIOSCIENCES INC., MISSISSAUGA, CA

Free format text: FORMER OWNER: LORUS THERAPEUTICS INC., TORONTO, ONTARIO, CA

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 931807

Country of ref document: AT

Kind code of ref document: T

Effective date: 20171015

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602004051855

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20180104

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171228

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171227

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: MICHELI AND CIE SA, CH

Ref country code: CH

Ref legal event code: PFA

Owner name: APTOSE BIOSCIENCES INC., CA

Free format text: FORMER OWNER: LORUS THERAPEUTICS INC., CA

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: APTOSE BIOSCIENCES INC.

REG Reference to a national code

Ref country code: BE

Ref legal event code: HC

Owner name: APTOSE BIOSCIENCES, INC.; CA

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), CHANGEMENT DE NOM DU PROPRIETAIRE, NOM/ADRESSE; FORMER OWNER NAME: LORUS THERAPEUTICS INC.

Effective date: 20180220

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602004051855

Country of ref document: DE

Owner name: APTOSE BIOSCIENCES INC., MISSISSAUGA, CA

Free format text: FORMER OWNER: LORUS THERAPEUTICS INC., TORONTO, ONTARIO, CA

REG Reference to a national code

Ref country code: NL

Ref legal event code: HC

Owner name: APTOSE BIOSCIENCES INC.; CA

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), CHANGE OF OWNER(S) NAME; FORMER OWNER NAME: LORUS THERAPEUTICS INC.

Effective date: 20180223

REG Reference to a national code

Ref country code: AT

Ref legal event code: PC

Ref document number: 931807

Country of ref document: AT

Kind code of ref document: T

Owner name: APTOSE BIOSCIENCES INC., CA

Effective date: 20180305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 14

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180127

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602004051855

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1094798

Country of ref document: HK

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171115

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

26N No opposition filed

Effective date: 20180628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20041115

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 931807

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170927

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20211126

Year of fee payment: 18

Ref country code: IE

Payment date: 20211129

Year of fee payment: 18

Ref country code: GB

Payment date: 20211129

Year of fee payment: 18

Ref country code: DK

Payment date: 20211129

Year of fee payment: 18

Ref country code: FR

Payment date: 20211124

Year of fee payment: 18

Ref country code: AT

Payment date: 20211020

Year of fee payment: 18

Ref country code: DE

Payment date: 20211126

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20211202

Year of fee payment: 18

Ref country code: BE

Payment date: 20211129

Year of fee payment: 18

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602004051855

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20221130

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20221201

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 931807

Country of ref document: AT

Kind code of ref document: T

Effective date: 20221115

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20221115

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20221130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221115

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221201

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221115

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221115

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230601

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130